Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Biochemistry

Cell Fractionation of U937 Cells by Isopycnic Density Gradient Purification

Published: August 12, 2021 doi: 10.3791/62119

ERRATUM NOTICE

Summary

This fractionation protocol will allow researchers to isolate cytoplasmic, nuclear, mitochondrial, and membrane proteins from mammalian cells. The latter two subcellular fractions are further purified via isopycnic density gradient.

Abstract

This protocol describes a method to obtain subcellular protein fractions from mammalian cells using a combination of detergents, mechanical lysis, and isopycnic density gradient centrifugation. The major advantage of this procedure is that it does not rely on the sole use of solubilizing detergents to obtain subcellular fractions. This makes it possible to separate the plasma membrane from other membrane-bound organelles of the cell. This procedure will facilitate the determination of protein localization in cells with a reproducible, scalable, and selective method. This method has been successfully used to isolate cytosolic, nuclear, mitochondrial, and plasma membrane proteins from the human monocyte cell line, U937. Although optimized for this cell line, this procedure may serve as a suitable starting point for the subcellular fractionation of other cell lines. Potential pitfalls of the procedure and how to avoid them are discussed as are alterations that may need to be considered for other cell lines.

Introduction

Subcellular fractionation is a procedure in which cells are lysed and separated into their constituent components through several methods. This technique can be used by researchers to determine protein localization in mammalian cells or for enrichment of low-abundance proteins that would otherwise be undetectable. While methods for subcellular fractionation currently exist, as do commercial kits that can be purchased, they suffer from several limitations that this procedure attempts to overcome. Most cell fractionation methods are exclusively detergent-based1,2, relying on the use of buffers containing increasing amounts of detergent to solubilize different cellular components. While this method is rapid and convenient, it results in impure fractions. These are designed to allow researchers to easily isolate one or two components of the cell, but are not complex enough to isolate multiple subcellular fractions from a sample at the same time. Relying solely on detergents usually results in membrane-enclosed organelles and the plasma membrane being indiscriminately solubilized, making separation of these components difficult. An additional complication from the use of these kits is the inability of researchers to alter/optimize them for specific applications, as most of the components are proprietary formulations. Finally, these kits can be prohibitively expensive, with limitations in the number of uses that make them less than ideal for larger samples.

Despite the availability of kits for the isolation of mitochondria that do not rely on detergents, they are not designed to isolate the plasma membrane and yield significantly lower amounts of sample than standard isolation protocols3,4. While differential centrifugation methods are more time-consuming, they often result in distinct fractions that cannot be obtained with exclusively detergent-based kits1. Separation without the sole use of solubilizing detergents also allows further purification using ultracentrifugation and isopycnic density gradients, resulting in less cross-contamination. This fractionation protocol demonstrates the isolation of subcellular fractions from U937 monocytes using a combination of detergent- and high-speed centrifugation-based approaches. This method will facilitate the isolation of the nuclear, cytoplasmic, mitochondrial, and plasma membrane components of a mammalian cell with minimal contamination between the fractions.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

1. Prepare buffers and reagents

  1. Prepare fresh solutions of phosphatase and protease inhibitors.
    1. Add 17.4 mg of phenylmethanesulfonyl fluoride (PMSF)to 1 mL of 100% ethanol to prepare a 100 mM stock.
      ​NOTE: Wear protective equipment when handling PMSF as it is hazardous when ingested or inhaled and upon contact with skin or eyes. It is corrosive to eyes and skin.
    2. According to the manufacturer's instructions, prepare a commercially available protease inhibitor cocktail (100x).
    3. Add 91.9 mg of sodium orthovanadate (SOV) to 1 mL of deionized water to prepare a 500 mM stock.
      NOTE: Wear protective equipment when handling SOV as it is hazardous if ingested, inhaled, or upon contact with eyes. Severe overexposure may result in death.
  2. Prepare lysis buffer A, cytoplasmic isolation (CI) buffer, cell solubilization (CS) buffer, nuclear lysis (NL) buffer, lysis buffer B, cell homogenization (CH) buffer, iodixanol diluent, and detergents.
    1. Add 8.7 g of sodium chloride (NaCl) and 50 mL of 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES, 1 M, pH 7.4) to 950 mL of deionized water to prepare lysis buffer A (final concentrations of 150 mM NaCl and 50 mM HEPES).
    2. Prepare stock solutions of detergents as follows: add 0.1 g of sodium dodecyl sulfate (SDS) to 10 mL of lysis buffer A (final concentration of 1% SDS), add 0.1 g of sodium deoxycholate to 10 mL of lysis buffer A (final concentration of 1%), add 2.5 mg of digitonin to 10 mL of lysis buffer A (final concentration of 250 µg/mL), and add 1 mL of non-ionic, non-denaturing detergent (see the Table of Materials) to 9 mL of lysis buffer A (final concentration of 10% (v/v)).
    3. Prepare CI buffer by adding 10 µL of PMSF stock (100 mM), 10 µL of protease inhibitor (100x), 2 µL of SOV stock (500 mM), and 100 µL of stock digitonin (250 µg/mL) to 878 µL of lysis buffer A (final concentrations of 1 mM PMSF, 1x protease inhibitor, 1 mM SOV, and 25 µg/mL digitonin). Keep the solution on ice until addition to cell pellet.
    4. Prepare CS buffer by adding 10 µL of PMSF stock (100 mM), 10 µL of protease inhibitor (100x), 2 µL of SOV stock (500 mM), 100 µL of non-ionic, non-denaturing detergent stock (see the Table of Materials) (10%), and 118 µL of hexylene glycol stock (8.44 M) to 760 µL of lysis buffer A (final concentrations of 1 mM PMSF, 1x protease inhibitor, 1 mM SOV, 1% non-ionic, non-denaturing detergent, and 1 M hexylene glycol). Keep the solution on ice until addition to cell pellet.
    5. Prepare NL buffer by adding 10 µL of PMSF stock (100 mM), 10 µL of protease inhibitor (100x), 2 µL of SOV stock (500 mM), 50 µL of sodium deoxycholate stock (10%), 100 µL of SDS stock (1%), and 118 µL of hexylene glycol stock (8.44 M) to 710 µL of lysis buffer A (final concentrations of 1 mM PMSF, 1x protease inhibitor, 1 mM SOV, 0.5% sodium deoxycholate (v/v), 0.1% SDS (w/v), and 1 M hexylene glycol). Keep the solution on ice until addition to nuclear pellet.
    6. Add 20 mL of HEPES (1 M, pH 7.4), 0.74 g of potassium chloride (KCl), 0.19 g of magnesium chloride (MgCl2), 2 mL of ethylenediaminetetraacetic acid (0.5 M EDTA), 2 mL of ethylene glycol-bis(β-aminoethyl ether)-N,N,N',N'-tetraacetic acid (0.5 M EGTA), 38.3 g of mannitol, and 23.9 g of sucrose to 980 mL of deionized water to prepare lysis buffer B (final concentrations of 20 mM HEPES, 10 mM KCl, 2 mM MgCl2, 1 mM EDTA, 1 mM EGTA, 210 mM mannitol, and 70 mM sucrose).
    7. Prepare CH buffer by adding 10 µL of PMSF stock (100 mM) and 2 µL of SOV stock (500 mM) to 988 µL of lysis buffer B (final concentrations of 1 mM PMSF and 1 mM SOV; adjust the final volume to accommodate the number of cells being lysed). Keep the solution on ice until addition to cell pellet.
    8. Prepare iodixanol diluent by addition of 12 mL of HEPES (1 M, pH 7.4), 447 mg of KCl, 114 g of MgCl2, 1.2 mL of 0.5 M EDTA, 1.2 mL of 0.5 M EGTA, 21.3 g of mannitol, and 14.4 g of sucrose to 88 mL of deionized water (final concentrations of 120 mM HEPES, 60 mM KCl, 12 mM MgCl2, 6 mM EDTA, 6 mM EGTA, 1200 mM mannitol, and 420 mM sucrose).
    9. Store buffers at 4 °C and digitonin at -20 °C.

2. Cytosolic protein isolation

NOTE: The following steps will allow for the growth and expansion of U937 cells followed by extraction of cytosolic proteins. At the concentration used, digitonin will permeabilize the plasma membrane without disrupting it, allowing for the release of cytosolic proteins and retention of other cellular proteins.

  1. Culture the cells in RPMI 1640 with 10% fetal bovine serum at 37 °C and 5% CO2. Ensure that the cells are grown to a final total of 6 x 108 cells.
    NOTE: In this protocol, cells were counted using a standard hemocytometer and microscope.
  2. Centrifuge the cultured cells at 400 × g for 10 min. After discarding the supernatant, resuspend the cell pellet in room temperature phosphate-buffered saline (PBS) at a final concentration of 4 × 106 cells/mL, and pipette gently to break up clumps.
  3. Centrifuge the cell suspension at 400 × g for 10 min to pellet the cells. Discard the supernatant, and resuspend the cell pellet in ice-cold lysis buffer A (prepared in step 1.2.1) at a final concentration of 2 × 107 cells/mL.
  4. Remove 7.5 mL of the cells suspended in lysis buffer A (3 × 107 cells), and keep on ice for nuclear protein extraction (section 6). Centrifuge the cell suspension at 4 °C, 400 × g for 10 min to pellet the cells.
    NOTE: Perform all subsequent steps at 4 °C or on ice, and prechill all buffers.
  5. Discard the supernatant, resuspend the cell pellet in CI buffer at a final concentration of 2 × 107 cells/mL, and pipette gently to break up clumps. Rotate the cell suspension end-over-end at 4 °C for 20 min.
  6. Centrifuge the cell suspension at 4 °C, 400 × g for 10 min, and transfer the supernatant to a clean centrifuge tube. Save the cell pellet, and store on ice. Centrifuge the collected supernatant at 4 °C, 18,000 × g for 20 min to pellet cellular debris.
    NOTE: This high-speed centrifugation step is critical for preventing contamination of the cytosolic fraction with organelle and membrane-bound proteins.
  7. Discard the pellet after transferring the supernatant to a clean centrifuge tube. Repeat both centrifugation steps in step 2.6 until no pellet is obtained following centrifugation.
  8. Collect the supernatant, which is the cytosolic fraction. For short-term storage (1 month), ensure that the supernatant is stored at 4 °C. For long-term storage (>1 month), combine the supernatant with 1x Laemmli buffer containing 1x reducing agent, heat at 95 °C for 7 min, and store at -20 or -80°C.
  9. Resuspend the cell pellet (from step 2.6) in lysis buffer A at a final concentration of 4 × 106 cells/mL; pipette gently to break up clumps.

3. Cell homogenization

NOTE: The following steps will allow for the mechanical homogenization of digitonin-treated cells (from step 2.9), which is necessary for the isolation of the mitochondrial and membrane protein fractions.

  1. Centrifuge the cell suspension in lysis buffer A (prepared in step 2.9), save the pellet, and discard the supernatant to remove excess digitonin and cytosolic contaminants from the cell pellet.
    NOTE: Repeated washes in lysis buffer A may be performed to remove excess cytosolic contaminants.
  2. Resuspend the cell pellet in ice-cold CH buffer (prepared in step 1.2.7) at a final concentration of 4 × 106 cells/mL. Incubate the cell suspension on ice for 30 min.
  3. If using a bead-based method for mechanical lysis, place 30 g of prewashed stainless steel 3.2 mm beads into a 50 mL skirted tube, fill with 15 mL of lysis buffer B, and place on ice to chill. If using a Dounce homogenizer (with a tight-fitting B pestle), fill with an appropriate volume of lysis buffer B, insert the pestle, and place on ice to chill.
  4. Following incubation (step 3.2), discard lysis buffer B from the bead tubes (or Dounce homogenizer), and transfer 15 mL of the cell suspension to the bead tube (or an appropriate volume to the homogenizer).
  5. If using the bead-based method, place the skirted bead tubes containing the cell suspension in the blender device, and configure to run for 5 min at speed 8. If using a Dounce homogenizer, keep it on ice, and perform 40 passes with the pestle using slow, even strokes (or utilize an alternative method of mechanical cell lysis as detailed in the discussion section).
    NOTE: If using a blender device different from the one used here, speed and time may need to be determined empirically.
  6. Transfer the homogenate to a clean centrifuge tube, and centrifuge it at 400 × g for 10 min at 4 °C. Transfer the supernatant to a clean centrifuge tube and save; discard the pellet.
    ​NOTE: The protocol may be paused here and the homogenate stored at 4 °C for the short term (24 h).

4. Debris removal and isolation of crude mitochondrial and membrane fractions

NOTE: The following steps will allow for the removal of cellular debris by centrifuging the homogenate at increasing speeds. This is followed by differential centrifugation for the isolation of crude mitochondrial and membrane fractions.

  1. Centrifuge the homogenate (from step 3.6) at 500 × g for 10 min at 4 °C. Transfer the supernatant to a clean centrifuge tube, and discard any pellet.
  2. Centrifuge the supernatant (from step 4.1) at 1,000 × g for 10 min at 4 °C. Transfer the supernatant to a clean centrifuge tube, and discard any pellet.
  3. Centrifuge the supernatant (from step 4.2) at 2,000 × g for 10 min at 4 °C. Transfer the supernatant to a clean centrifuge tube, and discard any pellet.
  4. Centrifuge the supernatant (from step 4.3) at 4,000 × g for 20 min at 4 °C. Transfer the supernatant to a clean centrifuge tube, and save the pellet, which is the crude mitochondrial fraction.
  5. Centrifuge the supernatant (from step 4.4) at 18,000 × g for 1 h at 4 °C. Discard the supernatant, and save the pellet, which is the crude membrane fraction.
    ​NOTE: The protocol may be paused here, and the pelleted samples stored at 4 °C for the short-term (24 h).

5. Isopycnic density gradient purification

NOTE: The following steps utilize isopycnic density gradient centrifugation to purify the crude mitochondrial and membrane fractions.

  1. Prepare a 50% (v/v) working solution of iodixanol by mixing 1 part diluent (prepared in step 1.1.3) to 5 parts iodixanol (60% stock solution (w/v)). Prepare 10%, 15%, 20%, 25%, 30%, and 35% iodixanol solutions (v/v) by mixing 50% working solution (v/v) with lysis buffer B in appropriate quantities.
  2. Resuspend the crude mitochondrial and membrane pellets (from steps 4.4 and 4.5) each in 200 µL of lysis buffer B. Adjust to 45% iodixanol (v/v) by adding 1800 µL of working iodixanol solution (50% (v/v)) to 200 µL of the resuspended pellet.
  3. Create an iodixanol discontinuous gradient as follows (adjust the volumes as appropriate for the capacity of the ultracentrifuge tube): Add 1 mL of 15% iodixanol (v/v) to the bottom of an 8 mL, open-top, thin-walled ultracentrifuge tube, place 1 mL of 20% iodixanol (v/v) below the first layer (by the underlaying technique), followed by underlaying 1 mL of 25%, 1 mL of 30%, and 1 mL of 35% iodixanol (v/v).
    NOTE: There should be 2 gradients created at this step, 1 for the mitochondrial fraction and 1 for the membrane fraction.
  4. In 1 gradient, add the 2 mL of the crude mitochondrial pellet (suspended in 45% iodixanol (v/v)) using the underlay technique to the bottom of the tube below the 35% iodixanol (v/v). In the other gradient, add the 2 mL of the crude membrane pellet (suspended in 45% iodixanol (v/v)) using the underlay technique to the bottom of the tube below the 35% iodixanol (v/v).
  5. Add 1 mL of 10% iodixanol (v/v) to the top of each gradient tube by overlaying it on top of the 15% layer. Balance the tubes within 0.1 g of each other by the addition of 10% iodixanol (v/v).
  6. Spin the density gradient tubes at 4 °C for 18 h at 100,000 × g. Be sure to set acceleration and deceleration to the minimum values for the ultracentrifuge being used.
    NOTE: In the mitochondrial gradient, there will be a visible band at the interface between 25% and 30% iodixanol (v/v). This is the pure mitochondrial fraction. In the membrane gradient, there will be a visible band in the 15% iodixanol (v/v) fraction. This is the pure membrane fraction. There may be additional bands in the 25% and 30% iodixanol (v/v) fractions in the membrane gradient. This is mitochondrial contamination.
  7. Collect fractions from the top of the tube in 1 mL aliquots, being careful to minimize the volume of the fraction containing the visible bands and changing the pipette tip after each layer is collected. Alternatively, puncture the side of the thin-walled tube with a needle, and collect the visible bands.
  8. Store the samples as described in step 2.8 until verification by protein assay and western blot.

6. Nuclear protein isolation

NOTE: Using ionic and non-ionic detergents as well as techniques such as sonication and centrifugation, the following steps will solubilize all cellular membranes and allow for the isolation of nuclear proteins.

  1. Centrifuge the 7.5 mL aliquot of cells suspended in lysis buffer A (from step 2.4), and discard the supernatant. Add 800 µL of ice-cold CS buffer (prepared in step 1.2.4), and resuspend the pellet by pipetting and vortexing. Incubate the samples on ice (or at 4 °C) for 30 min to disrupt the plasma and organelle membranes while protecting the nuclear proteins.
  2. Centrifuge the cell suspension at 7,000 × g for 10 min at 4 °C, and discard the supernatant. Add 800 µL of ice-cold NL buffer (prepared in step 1.2.5) to the nuclear pellet after inclusion of 1U/µL benzonase. Resuspend the pellet by gently pipetting. Incubate on an end-over-end rotator for 30 min at 4 °C to disrupt the nuclear membrane.
  3. Sonicate for 5 s at 20% power with sample tubes cooled in an ice bath (3x, with 5 s pauses in between pulses), which along with benzonase (added in step 6.2), will shear the nucleic acids.
  4. Centrifuge the sonicate at 7,800 × g for 10 min at 4 °C. Collect and save the supernatant, which is the nuclear fraction. Be sure to not disturb the insoluble pellet while collecting the supernatant. For storage, samples can be prepared as described in step 2.8.

7. Protein quantification and western blot analysis

NOTE: The following steps will quantify total protein in each fraction and confirm the purity of the subcellular fractions.

  1. Perform a standard Bradford assay to quantify total protein in each fraction. Refer to Table 1 for expected protein yields.
    NOTE: Other protein assays such as bicinchoninic acid (BCA) assay or absorbance at 280 nm may be used to measure total protein in place of a Bradford assay.
  2. Combine fractions with 1x Laemmli buffer containing 1x reducing agent, and heat at 95 °C for 7 min. Load 10 µg of each fraction into a standard SDS-polyacrylamide gel electrophoresis (PAGE) gel, and run the gel at 20 mA for 1 h.
  3. Transfer the resolved proteins to a polyvinyldifluoride (PVDF) membrane by performing a standard immunoblot transfer at 100 V for 30 min. Block the PVDF membrane with 5% milk in 1x Tris-buffered saline (TBS) containing 0.1% of a non-ionic detergent (v/v) for 30 min at room temperature.
  4. Add the appropriate primary antibodies to detect housekeeping proteins specific to each subcellular fraction, and incubate overnight at 4 °C. Wash the membrane with 5% milk in 1x TBS containing 0.1% of the non-ionic detergent (v/v) for 10 min at room temperature.
    NOTE: For this protocol, antibodies against glyceraldehyde-3-phosphate dehydrogenase (GAPDH, 1:10000), histone H3 (1:2000), voltage-dependent anion channel (VDAC, 1:1000), and Na,K+-ATPase were used for cytosolic, nuclear, mitochondrial, and membrane fractions, respectively.
  5. Add the appropriate horseradish peroxidase (HRP)-conjugated secondary antibodies to the membrane (dilute according to the manufacturer's instructions), and incubate at room temperature for 1 h. Wash the membrane with 5% milk in 1x TBS containing 0.1% of the non-ionic detergent (v/v) for 10 min at room temperature. Develop the blot using standard chemiluminescence.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

A schematic flow chart of this procedure (Figure 1) visually summarizes the steps that were taken to successfully fractionate U9375 cells grown in suspension. Fractions collected from the top of the isopycnic density gradient in equal volumes (1 mL) show the purification of the mitochondrial and membrane fractions (Figure 2). Utilizing an antibody against VDAC, a protein localized to the outer mitochondrial membrane6, shows that the mitochondrial fraction migrated to the 25% and 30% iodixanol (v/v) fractions (Figure 2A). Using an antibody against the Na,K+-ATPase α1 subunit, part of an integral membrane heterodimer found primarily in the plasma membrane7, shows the separation of membrane contamination from the pure mitochondrial fraction (Figure 2A). The pure membrane fraction migrated to the least dense fractions, 10% and 15% iodixanol (v/v) (Figure 2B). Mitochondrial contamination of the membrane fraction was separated by the gradient.

A western blot8 performed with the additional localization markers (referenced in step 7.4) shows the purity of the cytosolic and nuclear fractions, while additionally verifying that the mitochondrial and membrane samples are free from contamination by proteins from other parts of the cell (Figure 3). Using an antibody against GAPDH, normally localized to the cytoplasm of the cell9, shows that this protein is only found in the cytosolic fraction (Figure 3A, Lane 1, first panel), and that no contamination is observed in the extracted nuclear proteins, the density-purified mitochondria, or membrane fractions (Figure 3A; Lanes 2, 3, and 4; first panel). Probing for histone H3, a protein found in the nucleus and involved in chromatin structure10, shows a successful nuclear extraction (Figure 3A, Lane 2, second panel), with some minimal detection in the cytoplasmic fraction and no cross contamination in the mitochondrial or membrane fractions.

Probing for VDAC in all fractions shows the presence of this protein in the pure mitochondrial fraction (Figure 3A, Lane 3, third panel), and that no cross contamination exists in the other fractions (Figure 3A; Lanes 1, 2, and 4; third panel). Probing for the Na/K-ATPase α1 subunit similarly shows that this protein is located only in the pure membrane fraction (Figure 3A, Lane 4, fourth panel). These fractions were analyzed by densitometry to confirm reproducibility and statistical significance (Figure 3B-E). In contrast to the results from the successful fractionation (Figure 3), improper execution of this method (or failure to adhere to all recommended steps) can result in cross-contamination of cellular components (Figure 4). A high concentration of histone H3 in the cytosolic fraction (Figure 4, Lane 1, second panel) can result from a failure to properly clarify the cytosolic fraction (referenced in step 2.6). This can occur if not enough clarification centrifuge spins are performed, or if the cytosolic fraction is not clarified quickly. If the cytosolic fraction is not clarified quickly enough, it may result in lysis of cell fragments, leading to contamination of the cytosolic fraction.

Failure to perform the isopycnic density purification step will result in contamination of the membrane fraction (Figure 4, Lane 4, all panels), depending on how heterogenous the sample is prior to density purification. Proper adherence to all steps of the protocol is critical to obtaining the desired separation of the subcellular fractions. When quantified using a Bradford assay, protein yield for each fraction can be determined. Expected protein yield per fraction is reported in Table 1. It is useful to perform a protein quantification assay prior to performing western blots for several reasons. First, it confirms that fractions indeed contain protein; second, it allows loading of SDS-PAGE gels based on protein quantity; and finally, assuming protein yields are similar to those expected (Table 1), it confirms proper execution of the procedure.

Figure 1
Figure 1: Diagram of the cell fractionation procedure. An overview of the cell fractionation protocol represented as a flow chart. Abbreviations: PBS = phosphate-buffered saline; CS = cell solubilization; NL = nuclear lysis. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Isopycnic density purification of crude mitochondrial and membrane fractions. (A) Representative western blot of all fractions collected after density gradient purification of the crude mitochondrial fraction. (B) Representative western blot of all fractions collected after density gradient purification of the crude membrane fraction. Both density gradient purifications show migration of the mitochondrial marker, VDAC, for the 25% and 30% iodixanol (v/v) fractions and migration of the membrane marker, Na,K+ ATPase, for the 10% and 15% iodixanol (v/v) fractions. Abbreviation: VDAC = voltage-dependent anion channel. Please click here to view a larger version of this figure.

Figure 3
Figure 3: Successful isolation of U937 cytosolic, nuclear, mitochondrial, and membrane fractions. (A) Representative western blots of cell fractions isolated from a U937 cell culture with this technique and probed for markers of cytoplasm (GAPDH, first panel), nucleus (Histone H3, second panel), mitochondria (VDAC, third panel), and membrane (Na,K+ ATPase α1, fourth panel). (B-E) Densitometry of western blots of cell fractions isolated from U937 cell cultures with this technique. Results are from 3 independent experiments. Error bars represent standard deviation. One-way ANOVA. ***p<0.001. Abbreviations: GAPDH = glyceraldehyde-3-phosphate dehydrogenase; VDAC = voltage-dependent anion channel; cyto = cytosolic; nuc = nuclear; mito = mitochondrial; mem = membrane. Please click here to view a larger version of this figure.

Figure 4
Figure 4: Incomplete fractionation of U937 cell components. Representative western blots of cell fractions isolated from a U937 cell culture showing contamination of the cytosolic fraction with histone H3 (Lane 1, second panel) due to improper clarification of this fraction and a crude membrane fraction that was not subjected to isopycnic density gradient purification (Lane 4, all panels). Abbreviations: cyto = cytosolic; nuc = nuclear; mito = mitochondrial; mem = membrane. Please click here to view a larger version of this figure.

Fraction Yield (µg/mL) Standard Deviation Approximate Volume
Cytoplasm 1500 146 5 mL
Nucleus 1200 172 500 µL
Mitochondria 400 66 500 µL
Membrane 200 23 500 µL

Table 1: Protein yield for each subcellular fraction.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

This method is a modified version of a previously published approach to subcellular fractionation without the use of high-speed centrifugation11. This modified method requires more specialized equipment to achieve the best results, but is more comprehensive and consistently reproducible.

The development of the initial protocol was necessary due to an inability to separate mitochondrial and membrane samples for the analysis of protein localization during necroptosis12. Attempts to use the exclusively detergent-based methods found in most commercially available kits resulted in a homogeneous mixture containing the plasma membrane and all membrane-enclosed organelles in the cell. Other limitations of these kits include an inability to make alterations to the procedure, cost per sample, volume restrictions, and number of samples that can be processed. The procedure presented here may be altered to any scale, changed to isolate fewer fractions, and can be performed without the use of expensive reagents. Fraction yields can be increased by utilizing more cells; steps can be tailored to the research being performed; and execution of the method is flexible. For example, if researchers are not examining a particular subcellular fraction, they do not need to isolate that sample in the course of the procedure. Likewise, the addition of particular inhibitors or reagents can be omitted if the researcher does not plan on studying the phosphorylation state of proteins (sodium orthovanadate) or is not concerned about denaturing proteins (hexylene glycol).

The use of iodixanol as the density gradient solution is optional; however, this reagent does not interfere with subsequent examination of samples via western blotting. It is also possible to remove the iodaxanol from samples by dilution and centrifugation to recover the mitochondria or membrane, although this will affect the final yield. Other alternative density gradient solutions can be used, including sucrose. To obtain optimal results and pure fractions, there are several factors to consider, and particular critical steps in the protocol that need careful attention. This protocol is optimized for the fractionation of U937 cells, and the concentration of cells recommended at particular points in the protocol are specific to this cell line. These values were determined empirically and will most likely need to be adjusted for different types of cells, particularly, if suboptimal results are obtained when executing the protocol.

Clarification of the cytoplasmic sample should occur as soon as possible to remove unbroken cells and debris that might result in cross-contamination by proteins from other subcellular fractions (Figure 4, Lane 1, second panel). Homogenization can be accomplished with any form of mechanical lysis, although results presented here were obtained using a bead-based method and blender device. Alternative manual forms of homogenization (a Dounce homogenizer or passage through a small gauge needle) can also be utilized, but may result in reproducibility issues due to variability of technique by the individual performing the procedure. This group's interest is primarily in circulating leukocytes, which is why U937 cells are used in this procedure. However, this procedure may be applied to other suspension cell lines with likely little need for alteration. Portions that may need to be adjusted to accommodate another suspension cell line include cell concentrations used throughout the procedure as well as the concentration of digitonin used to extract the cytosolic fraction.

While not optimized for adherent cell lines, this procedure may serve as a starting point to which adjustments can be made to accommodate adherent cells. These adjustments include cell concentration, concentration of digitonin, and homogenization time. In addition, adherent cells are limited by surface area while suspension cells are limited by volume; this makes scaling up of suspension cells simpler. To scale up adherent cells, tissue culture plates with a large surface area (>500 cm2) must be utilized. This procedure is a cost-effective, reproducible subcellular fractionation with the ability to separate cytosolic, nuclear, mitochondrial, and membrane fractions with great purity. One of the biggest advantages of this procedure is the separation of mitochondrial from the membrane fraction. This is not possible in exclusively detergent-based procedures. Although detergents are used in this procedure, they are used to permeabilize, but not disrupt the plasma membrane (digitonin) and to obtain the nuclear fraction after the isolation of the mitochondrial and membrane fractions.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors declare no conflict of interest.

Acknowledgments

This work was supported by NIH R15-HL135675-01 and NIH 2 R15-HL135675-02 to T.J.L.

Materials

Name Company Catalog Number Comments
Benzonase Nuclease Sigma-Aldrich E1014
Bullet Blender Tissue Homogenizer Next Advance 61-BB50-DX
digitonin Sigma D141
end-over-end rotator ThermoFisher
Ethylenediaminetetraacetic acid (EDTA) Sigma E9884
ethylene glycol-bis(β-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA) Sigma E3889
GAPDH (14C10)  Cell Signalling Technologies 2118
HEPES VWR 97064-360
Hexylene glycol Sigma 68340
Igepal Sigma I7771 Non-ionic, non-denaturing detergent
KCl Sigma P9333
Mannitol Sigma M9647
MgCl2 Sigma M8266
NaCl Sigma S9888
Na, K-ATPase a1 (D4Y7E) Cell Signalling Technologies 23565
Open-Top Polyclear Tubes, 16 x 52 mm Seton Scientific 7048
OptiPrep (Iodixanol) Density Gradient Medium Sigma D1556-250ML
phenylmethanesulfonyl fluoride (PMSF) Sigma P7626
Protease Inhibitor Cocktail, General Use VWR M221-1ML
refrigerated centrifuge ThermoFisher
S50-ST Swinging Bucket Rotor Eppendorf
Sodium dodecyl sulfate (SDS) Sigma 436143
Sodium deoxycholate Sigma D6750
sodium orthovanadate (SOV) Sigma 567540
sonicator ThermoFisher
Sorvall MX120 Plus Micro-Ultracentrifuge ThermoFisher
Stainless Steel Beads 3.2 mm Next Advance SSB32
Sucrose Sigma S0389
Tris-buffered Saline (TBS) VWR 97062-370
Tween 20 non-ionic detergent in western blotting buffers
VDAC (D73D12) Cell Signalling Technologies 4661

DOWNLOAD MATERIALS LIST

References

  1. Baghirova, S., Hughes, B. G., Hendzel, M. J., Schulz, R. Sequential fractionation and isolation of subcellular proteins from tissue or cultured cells. MethodsX. 2, 440-445 (2015).
  2. Il Hwang, S., Han, D. K. Subcellular fractionation for identification of biomarkers: Serial detergent extraction by subcellular accessibility and solubility. Methods in Molecular Biology. 1002, 25-35 (2013).
  3. Clayton, D. A., Shadel, G. S. Isolation of mitochondria from cells and tissues. Cold Spring Harbor Protocols. 2014 (10), 1040-1041 (2014).
  4. Stimpson, S. E., Coorssen, J. R., Myers, S. J. Optimal isolation of mitochondria for proteomic analyses. Analytical Biochemistry. 475, 1-3 (2015).
  5. Sundström, C., Nilsson, K. Establishment and characterization of a human histiocytic lymphoma cell line (U-937). International Journal of Cancer. 17 (5), 565-577 (1976).
  6. Hodge, T., Colombini, M. Regulation of metabolite flux through voltage-gating of VDAC channels. Journal of Membrane Biology. 157 (3), 271-279 (1997).
  7. Therien, A. G., Blostein, R. Mechanisms of sodium pump regulation. American journal of physiology. Cell physiology. 279 (3), 541-566 (2000).
  8. Towbin, H., Staehelin, T., Gordon, J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proceedings of the National Academy of Sciences of the United States of the America. 76 (9), 4350-4354 (1979).
  9. Barber, R. D., Harmer, D. W., Coleman, R. A., Clark, B. J. GAPDH as a housekeeping gene: analysis of GAPDH mRNA expression in a panel of 72 human tissues. Physiological Genomics. 21 (3), 389-395 (2005).
  10. Bradbury, E. M., Cary, P. D., Crane-Robinson, C., Rattle, H. W. E. Conformations and interactions of histones and their role in chromosome structure. Annals of the New York Academy of Sciences. 222, 266-289 (1973).
  11. McCaig, W. D., Deragon, M. A., Haluska, R. J., Hodges, A. L., Patel, P. S., LaRocca, T. J. Cell fractionation of U937 cells in the absence of high-speed centrifugation. Journal of Visualized Experiments. (143), e59022 (2019).
  12. McCaig, W. D., et al. Hyperglycemia potentiates a shift from apoptosis to RIP1-dependent necroptosis. Cell Death Discovery. 4, 55 (2018).

Tags

Cell Fractionation U937 Cells Isopycnic Density Gradient Purification Cellular Compartments Cytoplasm Nucleus Mitochondria Membrane Scalable Procedure Reproducible Procedure Reliable Results Minimal Usage Of Detergents Cytosolic Protein Isolation RPMI 1640 Fetal Bovine Serum Centrifuge Cell Pellet PBS Lysis Buffer A Nuclear Protein Extraction Cytoplasmic Isolation Buffer

Erratum

Formal Correction: Erratum: Cell Fractionation of U937 Cells by Isopycnic Density Gradient Purification
Posted by JoVE Editors on 05/31/2022. Citeable Link.

An erratum was issued for: Cell Fractionation of U937 Cells by Isopycnic Density Gradient Purification. The Authors section was updated.

The Authors section was updated from:

William McCaig1
Timothy LaRocca1
1Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences

to:

William D. McCaig1
Matthew A. Deragon1
Phillip V. Truong1
Angeleigh R. Knapp1
Keven J. Hughes1
Timothy J. LaRocca1
1Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences

Formal Correction: Erratum: Cell Fractionation of U937 Cells by Isopycnic Density Gradient Purification
Posted by JoVE Editors on 07/22/2022. Citeable Link.

An erratum was issued for: Cell Fractionation of U937 Cells by Isopycnic Density Gradient Purification. The Authors section was updated.

The Authors section was updated from:

William D. McCaig1
Matthew A. Deragon1
Phillip V. Truong1
Angeleigh R. Knapp1
Keven J. Hughes1
Timothy J. LaRocca1
1Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences

to:

William D. McCaig1
Matthew A. Deragon1
Phillip V. Truong1
Angeleigh R. Knapp1
Timothy J. LaRocca1
1Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences

Cell Fractionation of U937 Cells by Isopycnic Density Gradient Purification
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

McCaig, W. D., Deragon, M. A.,More

McCaig, W. D., Deragon, M. A., Truong, P. V., Knapp, A. R., LaRocca, T. J. Cell Fractionation of U937 Cells by Isopycnic Density Gradient Purification. J. Vis. Exp. (174), e62119, doi:10.3791/62119 (2021).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter