Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Biology

In Vivo Detection and Analysis of Rb Protein SUMOylation in Human Cells

Published: November 2, 2017 doi: 10.3791/56096
* These authors contributed equally

Summary

Small ubiquitin-related modifier (SUMO) family proteins are conjugated to the lysine residues of target proteins to regulate various cellular processes. This paper describes a protocol for the detection of retinoblastoma (Rb) protein SUMOylation under endogenous and exogenous conditions in human cells.

Abstract

The post-translational modifications of proteins are critical for the proper regulation of intracellular signal transduction. Among these modifications, small ubiquitin-related modifier (SUMO) is a ubiquitin-like protein that is covalently attached to the lysine residues of a variety of target proteins to regulate cellular processes, such as gene transcription, DNA repair, protein interaction and degradation, subcellular transport, and signal transduction. The most common approach to detecting protein SUMOylation is based on the expression and purification of recombinant tagged proteins in bacteria, allowing for an in vitro biochemical reaction which is simple and suitable for addressing mechanistic questions. However, due to the complexity of the process of SUMOylation in vivo, it is more challenging to detect and analyze protein SUMOylation in cells, especially when under endogenous conditions. A recent study by the authors of this paper revealed that endogenous retinoblastoma (Rb) protein, a tumor suppressor that is vital to the negative regulation of the cell cycle progression, is specifically SUMOylated at the early G1 phase. This paper describes a protocol for the detection and analysis of Rb SUMOylation under both endogenous and exogenous conditions in human cells. This protocol is appropriate for the phenotypical and functional investigation of the SUMO-modification of Rb, as well as many other SUMO-targeted proteins, in human cells.

Introduction

The accurate control of cell cycle progression in eukaryotic cells is based on a tight regulatory network, which ensures that particular events take place in an ordered manner1,2. One of the key players in this network is the retinoblastoma (Rb) protein, the first cloned tumor suppressor1,3. The Rb protein is thought to be a negative regulator of cell cycle progression, especially for the G0/G1 to S phase transition, and tumor growth4,5. Failure of Rb function either directly leads to the most common intraocular malignancy in children, retinoblastoma, or contributes to the development of many other types of cancer5. Moreover, Rb is involved in many cellular pathways including cell differentiation, chromatin remodeling, and mitochondria-mediated apoptosis3,6,7.

Post-translational modifications play a pivotal role in the regulation of RB function8,9. Phosphorylation is one such modification, and it usually leads to Rb inactivation. In quiescent G0 cells, Rb is active with a low phosphorylation level. As cells progress through G0/G1 phase, Rb is sequentially hyper-phosphorylated by a series of cyclin-dependent protein kinases (CDKs) and cyclins, such as cyclin E/CDK2 and cyclin D/CDK4/6, which inactivate Rb and eliminate its ability to repress cell-cycle related gene expression4,10. Rb could also be modified by small ubiquitin-related modifier (SUMO)11,12,13.

SUMO is a ubiquitin-like protein that is covalently attached to a variety of target proteins. It is crucial for diverse cellular processes, including cell cycle regulation, transcription, protein cellular localization and degradation, transport, and DNA repair14,15,16,17,18. The SUMO conjugation pathway consists of the dimeric SUMO E1 activating enzyme SAE1/UBA2, the single E2 conjugating enzyme Ubc9, multiple E3 ligases, and SUMO-specific proteases. Generally, nascent SUMO proteins must be proteolytically processed to generate the mature form. The mature SUMO is activated by the E1 heterodimer and then transferred to the E2 enzyme Ubc9. Finally, the C-terminal glycine of SUMO is covalently conjugated to the target lysine of a substrate, and this process is usually facilitated by E3 ligases. The SUMO protein can be removed from the modified substrate by specific proteases. A previous study by the authors of this paper revealed that SUMOylation of Rb increases its binding to CDK2, leading to hyper-phosphorylation at the early G1 phase, a process which is necessary for cell cycle progression13. We also demonstrated that the loss of Rb SUMOylation causes a decreased cell proliferation. Moreover, it was recently demonstrated that the SUMOylation of Rb protects the Rb protein from proteasomal turnover, thus increasing the level of Rb protein in cells19. Therefore, SUMOylation plays an important role in Rb function in various cellular processes. To further study the functional consequence and physiological relevance of Rb SUMOylation, it is important to develop an effective method to analyze the SUMO status of Rb in human cells or patient tissues.

SUMOylation is a reversible, highly dynamic process. Thus, it is usually difficult to detect the SUMO-modified proteins under completely endogenous conditions. This paper presents a method to detect endogenous Rb SUMOylation. Furthermore, it shows how to detect exogenous Rb SUMOylation of both wild-type Rb and its SUMO-deficient mutation11. In particular, Jacobs et al. described a method to increase the SUMO modification of a given substrate specifically by Ubc9 fusion-directed SUMOylation (UFDS)20. Based on this method, this protocol describes how to analyze the forced SUMOylation of Rb and its functional consequences. Given that hundreds of SUMO substrates have been described previously and more putative SUMO substrates have been identified from many proteomic-based assays, this protocol can be applied to analyze the SUMO-modification of these proteins in human cells.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

1. Detection of Endogenous Rb SUMOylation at the Early G1 Phase

  1. Cell culture and cell cycle synchronization.
    1. Maintain HEK293 cells in growth medium containing Dulbecco's Modified Eagle Medium (DMEM) supplemented with 1% Pen-Strep and 10% fetal bovine serum (FBS) at 37 °C and 5% CO2 in an incubator.
    2. Synchronize the HEK293 cells at the G0 phase.
      1. Count the HEK293 cells using a hemocytometer and seed ~1.5 x 107 cells in a 15 cm dish with 25ml growth medium for 24 h before treatment. After reaching a confluence of 70% - 80%, aspirate the medium and wash the cells twice with 5 mL prewarmed phosphate-buffered saline (PBS).
      2. Add 25 mL DMEM containing 1% Penicillin-Streptomycin and incubate the cells at 37 °C for 72 h. Wash the cells off the plate using 3 mL ice-cold PBS. Transfer the cells into a new 5 mL tube.
      3. Subject a small portion of cells to cell cycle analysis by flow cytometry (step 1.2); collecting the remaining majority of the cells by centrifugation at 200 x g for 5 min at room temperature, and then store them in an ultra-lower temperature freezer until analysis of Rb SUMOylation.
    3. To obtain G1-phase cells, at the end of the G0 synchronization process, remove the DMEM and add back fresh growth medium, thus allowing the HEK293 cells to re-enter the cell cycle. Then, collect the cells at different G1 phases (early G1: 30 min; G1: 2h) for cell cycle analysis and further SUMO assay.
    4. Synchronize the HEK293 cells at the S phase using the double-thymidine block method.
      1. Grow HEK293 cells to a confluency of 50% and wash cells with prewarmed PBS. Then add growth medium supplemented with 2.5 mM thymidine for 18 h (first block).
      2. Remove the thymidine-containing medium, and then wash the cells twice with prewarmed PBS. Add fresh growth medium for 14 h to release the cells.
      3. Discard the medium with a pipette and add growth medium supplemented with 2.5 mM thymidine for another 18 h (second block) before conducting an analysis of the cell cycle and Rb SUMOylation.
    5. For the G2/M synchronization, plate ~1.5 × 107 HEK293 cells to a 15 cm dish with 25 mL growth medium for 24 h. Then add nocodazole to the medium until a final concentration of 400 ng/mL is obtained. Finally, incubate the cells for 16 h before conducting the analysis of the cell cycle and Rb SUMOylation.
  2. Cell cycle analysis by flow cytometry.
    1. Re-suspend the synchronized HEK293 in PBS, and then fix the cell suspension using ice-cold 70% ethanol for 2 h at 4 °C. Note that to minimize cell clustering, add the cell suspension dropwise to the ice-cold 100% ethanol to obtain a final concentration of 70% ethanol while gently vortexing.
    2. Centrifuge the cells for 5 min at 500 x g, then carefully discard the supernatant and wash the cells twice with PBS. Repeat the centrifuge step.
    3. Add 500 µL PBS containing 50 µg/mL nucleic acid stain propidium iodide, 0.1% Triton X-100 and 1 µg/mL RNase A to the cells and mix well. Incubate the cells for 15 min at 37 °C.
    4. Store the samples at 4 °C until analysis by flow cytometry.
  3. Immunoprecipitation of endogenous Rb protein.
    1. Prepare the HEK293 cell lysates.
      1. Lyse the synchronized HEK293 cells by gently re-suspending them in 1 mL of ice-cold radio-immunoprecipitation assay (RIPA) lysis buffer (Table 1) containing freshly added 20 mM N-Ethylmaleimide, an isopeptidase inhibitor that could block SUMO proteases and stabilize SUMO conjugates.
      2. Further homogenize the cells on ice by Dounce homogenization, sonication or simply passing through 10 times through a 21 G needle attached on a 2 mL syringe. Then, incubate the cells on ice for 5 min.
        NOTE: The anionic detergent sodium dodecyl sulfate (SDS) in the RIPA buffer is crucial for the later determination of the Rb-SUMO, as it could eliminate the unspecific SUMO signal that is derived from the non-covalent interaction between the Rb protein and other non-Rb SUMO-species.
    2. Centrifuge the cell lysates at 18,000 x g for 30 min at 4 °C. Transfer the supernatants to new 1.5 mL micro-centrifuge tubes.
      NOTE: The protocol can be paused here. The proteins can be stored at -80 °C for at least 6 months.
    3. To prepare the input control of each sample for the later Western blot, save a small portion of the above-described supernatant to a new tube and store at -80 °C.
    4. To the above supernatants, add 1 µg of non-specific mouse immunoglobulin G (IgG) of the same species and isotype as the monoclonal Rb antibody, and 20 µL of 50% protein A/G-sepharose slurry. Then, incubate for 1 h at 4 °C with gentle rotation.
    5. Centrifuge the samples at 3,000 x g for 3 min at 4 °C. Carefully collect the supernatants without disturbing the beads, and transfer them to new 1.5 mL micro-centrifuge tubes. To each of the samples, add 5 µL Rb primary antibody and 40 µL of 50% protein A/G-sepharose slurry. Then, incubate overnight at 4 °C with gentle rotation.
    6. Collect the beads by centrifugation at 3,000 x g for 3 min at 4 °C, and carefully remove the supernatant by pipetting. Note that in order to avoid bead loss, do not aspirate the beads dry.
    7. Wash the beads four times with 1 mL of the RIPA buffer. Each time, mix the tubes well by rotation them at 4 °C for 15 min. Collect the beads by low speed centrifugation at 3,000 x g for 3 min at 4 °C and then discard the supernatants.
    8. After removing supernatants from the final wash, re-suspend the beads in 30 µL 1x sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) loading buffer (Table 1) and mix well.
      1. Incubate the tubes at 100 °C in a heat block for 10 min. Centrifuge the samples at 12,000 x g for 1 min to pellet the beads. Carefully collect the supernatants without disturbing the beads, and transfer them to 1.5 mL micro-centrifuge tubes.
    9. Analyze the samples by 4% - 20% gradient SDS-PAGE gel and Western blot or store them at -20 °C for later use.

2. Analysis of 6XHis-tagged Exogenous Rb SUMOylation in Human Cells

  1. Cell culture and transfection.
    1. Seed ~6 × 106 HEK293 cells in a 10 cm dish and incubate for 24 h in growth medium under normal cell culture conditions to acquire 75%-85% confluent cells. Before transfection, remove the growth medium and add 6 mL of prewarmed reduced serum medium (see Table of Materials) and then place the dishes back into the incubator.
    2. For the Rb constitutive SUMOylation assay, add 15 µg each of the 6XHis-tagged Rb-Ubc9-WT, Rb-Ubc9-C93S, Ubc9-WT and Ubc9-C93S plasmids in 500 µL reduced serum medium in 1.5 mL micro-centrifuge tubes, respectively.
      1. To analyze the SUMO-conjugation of wild type Rb and its SUMOylation-defective mutant, mix 10 µg of either 6XHis-tagged Rb-WT or Rb-K720R plasmid together with GFP-SUMO1 (10 µg) in 500 µL reduced serum medium in 1.5 mL micro-centrifuge tubes13.
    3. Meanwhile, in a separate tube, mix 50 µL transfection reagent (see Table of Materials) in 500 µL reduced serum medium, and incubate at room temperature for 5 min.
    4. Fully combine the two separate tubes described above, and incubate at room temperature for 15 min. Add the transfection reagent/plasmid complexes to the cells and continue to culture for 8 h at 5% CO2 and 37 °C.
    5. Replace reduced serum medium with growth medium, and incubate the cells under normal culture conditions for 48 h after transfection.
  2. Nickel affinity pull down of the 6XHis-tagged Rb protein.
    1. Lyse the transfected HEK293 cells and extract the total proteins as described Section 1.3.1.
    2. Centrifuge the cell lysates at 18,000 x g for 30 min at 4 °C. Carefully collect the supernatants and transfer them to clean tubes.
      NOTE: The protein can be frozen at this point for more than 6 months at -80 °C.
    3. To prepare the input control of each sample for the later Western blot, save a small portion of the supernatants described above to a new tube and store at -80 °C.
    4. Wash 25 µL of 50% nickel nitrilotriacetic acid (Ni-NTA) agarose beads with RIPA buffer twice in a 1.5 mL micro-centrifuge tube for each sample. Collect the beads by centrifugation at 3,000 x g for 3 min at 4 °C.
    5. Add 1 M imidazole to each sample to obtain a final concentration of 10 mM. Then, add each sample to the tube containing the prepared Ni-NTA agarose beads.
    6. Incubate the beads and the lysates for 2 h at 4 °C with gentle rotation. Spin the beads at 3,000 x g for 3 min at 4 °C, and carefully remove the supernatants by pipetting. To avoid bead loss, do not aspirate the beads dry.
    7. Wash the beads with 1 mL of wash buffer containing 20 mM imidazole (Table 1). Each time, mix the tubes well by rotation at 4 °C for 15 min. Collect the beads by spinning at 3,000 x g for 3 min at 4 °C, and discard the supernatants.
    8. After the final wash, add 30 µL of elution buffer containing 250 mM imidazole (Table 1) to each sample and flick to mix. Then, incubate for 20 min at 4 °C to elute the proteins.
    9. Mix each sample with 6× SDS-PAGE loading buffer (Table 1). Then, incubate the tubes at 100 °C in a heat block for 10 min.
    10. Centrifuge at 12,000 x g for 1 min to pellet the beads. Carefully collect the supernatants without disturbing the beads, and transfer the samples to 1.5 ml micro-centrifuge tubes. The samples can be analyzed by Western blot or stored at -20 °C for later use.

3. Western Blot

  1. Load the immunoprecipitation or pull down samples obtained from the previous steps onto 4-20% gradient SDS-PAGE gels. Conduct electrophoresis at 120 V for 90 min to separate the proteins.
  2. Transfer the proteins from the gel to a polyvinylidene difluoride (PVDF) membrane by electroblotting at 300 mA for 90 min at 4 °C using the tank transfer method. Block the PVDF membrane in block buffer containing 5% nonfat milk (Table 1) for 1 h at room temperature.
  3. Incubate the membrane with primary antibody in antibody dilution buffer containing 3% bovine serum albumin (BSA) (Table 1) overnight at 4 °C. For primary antibodies, use a working concentration of 0.5 µg/mL (anti-SUMO1 antibody), or a dilution of 1:2000 (anti-Rb and anti-Tubulin antibodies), or 1:5,000 (anti-GFP and anti-His antibody).
  4. Wash the membrane 3x for 10 min each time using 1x tris-buffered saline with Tween (TBST) buffer (Table 1). Incubate the membrane with species specific Horseradish Peroxidase (HRP)-conjugated secondary antibodies diluted 1:5,000 in block buffer containing 5% nonfat milk (Table 1). Wash the membrane 3x for 10 min each time using 1x TBST buffer.
  5. Incubate the membrane with enhanced chemiluminescence (ECL) working solution. Cover the membrane with plastic wrap and expose it to X-ray film depending on the strength of signal.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

To detect endogenous Rb SUMOylation during cell cycle progression, this study first synchronized HEK293 cells at five different stages of the cell cycle (G0, early G1, G1, S, and G2/M) as described in the protocol section of this paper. The quality of synchronization was confirmed by using the nucleic acid stain with propidium iodide followed by flow cytometry analysis (Figure 1). Next, the cells were collected and lysed by denaturing RIPA buffer.The SUMO proteases inhibitor, N-Ethylmaleimide, was added to a final concentration of 20 mM to preserve the native SUMO signal during the experiments. After immunoprecipitation of the endogenous Rb species under denaturing conditions to block the non-covalent unspecific interaction, and the following western blot using an anti-SUMO1 antibody, the presence of the SUMO signal was specifically detected at the early G1 phase (Figure 2). Although the global SUMOylation was enhanced at the S/G2/M phase, at the time point of Rb SUMOylation, it had not changed (Figure 2, input panel). This finding suggests that the SUMOylation of Rb is not simply the consequence of altered global SUMO conjugation activity. Thus, these results show that the protein immunoprecipitation and Western blot analysis described in this paper allow for the detection of SUMOylation of the endogenous Rb protein.

To detect the forced SUMOylation of the Rb protein, this study generated a constitutive SUMOylated Rb construct by fusing Ubc9, the sole SUMO E2 ligase, to its C-terminal allowing for efficient and selective SUMOylation of Rb (Figure 3A). The loss-of-function mutation of Ubc9, C93S, is also fused to the C-terminal of Rb to serve as a control (Figure 3A). To further strengthen the specificity of the SUMO-Rb signal, non-fused Ubc9 alone was constructed as well. All four of the His-tagged plasmids were transfected into the HEK293 cells for 48 h before they were lysed in RIPA buffer supplemented with 20 mM N-Ethylmaleimide. All the proteins were then subjected to a pull down assay, as described in the protocol section of this paper. The eluted Rb proteins were analyzed by Western blot. The Ubc9 fusion-directed SUMOylation of Rb was detected using an anti-SUMO1 antibody (Figure 3B, SUMO1 panel), whereas the Ubc9 defective mutation, C93S, failed to produce this signal. Note that the highly efficient SUMO-modification caused by Ubc9-fusion leads to a higher molecular weight band that could even be directly detected by using the Rb antibody, and it corresponds to the SUMO signal (Figure 3B, Rb panel). Moreover, Ubc9 alone did not cause any SUMO conjugation, further confirming the Rb-specific SUMOylation (Figure 3B).

Because SUMO1 is conjugated to lysine 720 of the Rb protein11, to further analyze the Rb SUMOylation, this study generated a SUMO-deficient mutation by replacing this lysine residue with an arginine (K720R). To facilitate the detection of SUMO-conjugation of the two transiently expressed Rb proteins, a GFP-SUMO1 construct was added to enhance the intracellular SUMO signal. His-tagged wild type or mutant Rb was co-transfected into HEK293 cells together with GFP-SUMO1, followed by pull down assay and analysis of Rb-SUMO1 conjugation capability. As observed, the K720R mutant totally abolished the SUMO-modification of Rb, further strengthening the proposed method's ability to detect the Rb SUMOylation (Figure 4).

Figure 1
Figure 1: Validation of the cell cycle synchronization assay by flow cytometry. HEK293 cells were cultured and synchronized at the G0, early G1, G1, S, and G2/M phases of the cell cycle as described in this protocol. Cell cycle distributions were determined by fluorescence-activated cell sorting (FACS). The represented data shows an example of the quantitative analysis of the FACS assay (n = 1). Please click here to view a larger version of this figure.

Figure 2
Figure 2: Rb is SUMOylated at early G1 phase. HEK293 cells were synchronized at different phases of the cell cycle as described in this protocol. The cells were collected and lysed in RIPA buffer supplemented with 20 mM N-Ethylmaleimide. The input controls were loaded on a 4% - 20% SDS-PAGE gradient gel, transferred, and blotted with anti-SUMO1 and anti-Tubulin, as indicated. The remaining cell lysates were then subjected to immunoprecipitation using anti-Rb antibody at a dilution of 1:200. The resulting eluents were separated by 4% - 20% SDS-PAGE gradient gel and immunoblotted using anti-SUMO1 antibody and anti-Rb antibody. This experiment was repeated twice with the same result. This figure has been modified with permission13. Please click here to view a larger version of this figure.

Figure 3
Figure 3. Detection of the SUMO-modification of Rb-Ubc9 fusion protein. (A) Diagram of the Ubc9 fusion-directed SUMOylation (UFDS) constructs of Rb. (B) Constitutive SUMOylation of Rb caused by UFDS. HEK293 cells transiently transfected with His-tagged UFDS constructs were lysed in RIPA buffer with 20 mM N-Ethylmaleimide. The total lysate of each sample was incubated with Ni-NTA agarose beads to pull down the His-tagged Rb-Ubc9 fusion proteins or Ubc9 (used as the negative control), respectively. The purified proteins were separated by 4% - 20% SDS-PAGE gradient gel and immunoblotted with anti-SUMO1 and anti-His antibodies. His-Rb: 6XHis tagged Rb-Ubc9 fusion proteins; His-Ubc9: 6XHis tagged Ubc9 only. Rb-Ubc9: unmodified Rb-Ubc9; Rb-Ubc9-S: SUMOylated Rb-Ubc9; Hyper-pRb-Ubc9: hyper-phosphorylated Rb-Ubc9. The results shown in the figure are representative of three independent trials. This figure has been modified with permission13. Please click here to view a larger version of this figure.

Figure 4
Figure 4. The SUMO-deficient K720R mutation reduces the SUMOylation of Rb. HEK293 cells were transiently co-transfected with the wild type or mutant Rb-His constructs together with GFP-SUMO1. The cells were collected and lysed in RIPA buffer supplemented with 20 mM N-Ethylmaleimide. A small portion of the lysates was directly analyzed by Western blot as the input control. The rest of the lysates were incubated with Ni-NTA agarose beads to pull down the His-tagged Rb proteins as described in this protocol, and they were immunoblotted with anti-SUMO1 and anti-His antibodies. Note that the lysine-to-arginine mutation at 720 of Rb totally abolishes the SUMO modification of this protein. The experiment was conducted once with a similar result to that previously reported11. This figure has been modified with permission13. Please click here to view a larger version of this figure.

Solution Components Comments
RIPA Lysis Buffer 50 mM Tris, pH = 8.0; 150 mM NaCl; 1% NP-40; 1% sodium deoxycholate; 0.1% SDS Add protease inhibitor cocktail and N-ethylmaleimide immediately before use.
Wash Buffer 50 mM NaH2PO4 pH = 8.0; 300 mM NaCl; 20 mM imidazole Used for Pull down assay only
Elution Buffer 50 mM NaH2PO4 pH = 8.0; 300 mM NaCl; 250 mM imidazole  Used for Pull down assay only
6x Loading Buffer 0.5 M Tris, pH = 6.8; 30% glycerol; 10% SDS; 5% β-mercaptoethanol; 0.1% bromphenol blue  Store at -20 °C
10x Running Buffer 0.25M Tris, pH 8.6; 1.9M Glycine; 1% SDS To make 1x Running Buffer: mix 100 ml 10x Running Buffer with 900 mL ddH2O
10x Transfer Buffer 0.25 M Tris, pH 8.3, 1.9 M Glycine  To make 1x Transfer Buffer: mix 100 mL 10x Transfer Buffer with 200 ml Methanol and 700 mL ddH2O
10x TBS Buffer In 1 L of ddH2O: 24.08 g Tris pH 7.4; 80 g NaCl To make 1x TBST Buffer: mix 100 mL 10x TBS Buffer with 900 mL ddH2O and 1 mL of Tween-20
Blocking Buffer 1x TBST with 5%  nonfat dry milk Prepare the buffer immediately before use
Antibody Dilution Buffer 1x TBST with 3% BSA  and 0.02% sodium azide This Buffer could be stored at 4 °C for at least 6 month

Table 1: Solutions and buffers.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

This paper describes a protocol to detect and analyze the endogenous SUMOylation of Rb in human cells. As this method is specifically focused on the endogenous Rb protein without any alternation of global SUMO-related signal, it is an important tool for investigating Rb-SUMO modification under completely natural physiologic circumstances.

To achieve this aim, it is important to keep in mind that: 1) although SUMO comprises four isoforms (SUMO1-4, each encoded by different genes)in comparison to ubiquitination, all the SUMO species are much less abundant; 2) for most SUMO target proteins, only a small portion of a given protein is SUMOylated at steady state, which is the outcome of the constant competition between the enzymes involved in SUMO conjugation and deconjugation14,21; and 3) SUMO targets can undergo rapid cycles of SUMOylation and deSUMOylation. For example, recent data obtained by the authors of this current paper demonstrate that Rb-SUMO1 exists only for a very short window of time in the early G1 phase, which is consistent with the conception that SUMOylation is a reversible, highly dynamic process13. All these facts make it challenging to directly detect the endogenous SUMOylation of a given protein. Thus, to successfully detect this, it is vital to identify the exact conditions under which this modification occurs. For instance, in this proposed protocol, the timing of the cell cycle synchronization is crucial for the detection of Rb SUMOylation. An optimized experimental procedure is also important for the successful detection of the low-level SUMO-modified species of a given protein. For example, proper sonication or passing-through needles could prevent the formation of sticky and viscous components due to genomic DNA, thus proper sonication can facilitate total protein extraction. Moreover, a good monoclonal antibody with high affinity to the target protein could be helpful for improving the quantity and specificity of immunoprecipitation. In summary, the proposed method is important for further exploring the physiological conditions under which endogenous Rb is SUMOylated, which is the premise of the following overexpression-based functional assay.

To amplify the SUMO signal of a given protein, it is common to co-overexpress this protein as well as other SUMO-related proteins (usually Ubc9 and SUMO protein) in cells. Although a simple Western blot using an antibody against the substrate is the easiest way to find the higher molecular weight, SUMOylated form of this protein, we failed to detect the SUMO-Rb signal with this method. Thus, this paper only focused on a method to detect the SUMO-modification of the precipitated exogenous Rb protein. Moreover, this method described how to artificially enhance or eliminate the SUMO-conjugation of Rb. As the sole E2 ligase, Ubc9 has been found to directly transfer SUMO to specific targets22. Thus, by fusing to the C terminal of Rb, Ubc9 significantly promotes the SUMO-modification of Rb. Using this method, the authors of this paper successfully demonstrated that SUMOylation of Rb sufficiently promoted its own phosphorylation by increasing its binding to CDK213. Moreover, to study the functional consequences of the loss of Rb SUMOylation, the authors generated a SUMO-deficient Rb construct, as previously reported11. By using the method proposed in this current paper, it was shown that SUMO-Rb is necessary for normal cell cycle progression and cell proliferation13.

In addition to SUMO1, SUMO2 and SUMO3 also play important roles in protein SUMOylation14,17. SUMO2 and SUMO3 are often referred to as SUMO2/3 because they are closely related and share 97% identity. SUMO1 shares a 50% similarity with SUMO2/3. It is widely accepted that SUMO1 is responsible for normal cell physiological functions and maintenance, whereas SUMO-2/3 is predominantly involved in cell stress responses15,17,23,24. Given that Rb could also be SUMOylated by SUMO2/3 with unknown function12, the method proposed in this study is still suitable for the detection and analysis of this modification of Rb. In addition to Rb, the methods described here can be widely applied to the detection and functional analysis of the SUMOylation of a variety of target proteins.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have nothing to disclose.

Acknowledgments

This study was supported by grants from the Science and Technology Commission of Shanghai (Grant No. 14411961800) and National Natural Science Foundation of China (Grant No. 81300805).

Materials

Name Company Catalog Number Comments
Dulbecco's Modified Eagle Medium (DMEM) Thermo Fisher Scientific 11995065
Opti-MEM  Thermo Fisher Scientific 31985070
Fetal Bovine Serum (FBS) Thermo Fisher Scientific 26140079
Penicillin-Streptomycin Thermo Fisher Scientific 15140122
Phosphate-buffered Saline (PBS) Thermo Fisher Scientific 10010023
Trypsin-EDTA Thermo Fisher Scientific 25200056
Thymidine Sigma T9250
Nocodazole Sigma M1404
propidium iodide Thermo Fisher Scientific P3566
Triton X-100  AMRESCO 694
RNase A  Thermo Fisher Scientific EN0531
N-Ethylmaleimide Sigma E3876 
Sodium Dodecyl Sulfate (SDS) AMRESCO M107
Nonidet P-40 Substitute (NP-40) AMRESCO M158
protease inhibitor Roche 5892970001
Mouse Immunoglobulin G (IgG) Santa Cruz Biotechnology sc-2025
Rb antibody Cell Signaling Technology #9309
Protein A/G-Sepharose Beads Santa Cruz Biotechnology sc-2003
Lipofectamine-2000  Thermo Fisher Scientific 11668019
Nickel Nitrilotriacetic Acid (Ni-NTA) Agarose Beads Qiagen 30230
Imidazole Sigma I0250
4%-20% Gradient SDS-PAGE Gel BIO-RAD 4561096
Polyvinylidene Difluoride (PVDF) Membrane Millipore IPVH00010
Tween-20 AMRESCO M147
Tubulin antibody Abmart M30109
SUMO1 antibody Thermo Fisher Scientific 33-2044
GFP antibody Abmart M20004
Horseradish Peroxidase (HRP) secondary antibody Jackson ImmunoResearch Laboratories 715-035-150
enhanced chemiluminescence (ECL) Kit Thermo Fisher Scientific 32106

DOWNLOAD MATERIALS LIST

References

  1. Bertoli, C., Skotheim, J. M., de Bruin, R. A. Control of cell cycle transcription during G1 and S phases. Nat Rev Mol Cell Biol. 14 (8), 518-528 (2013).
  2. Massague, J. G1 cell-cycle control and cancer. Nature. 432 (7015), 298-306 (2004).
  3. Weinberg, R. A. The retinoblastoma protein and cell cycle control. Cell. 81 (3), 323-330 (1995).
  4. Giacinti, C., Giordano, A. RB and cell cycle progression. Oncogene. 25 (38), 5220-5227 (2006).
  5. Burkhart, D. L., Sage, J. Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat Rev Cancer. 8 (9), 671-682 (2008).
  6. Ferreira, R., Naguibneva, I., Pritchard, L. L., Ait-Si-Ali, S., Harel-Bellan, A. The Rb/chromatin connection and epigenetic control: opinion. Oncogene. 20 (24), 3128-3133 (2001).
  7. Hilgendorf, K. I., et al. The retinoblastoma protein induces apoptosis directly at the mitochondria. Genes Dev. 27 (9), 1003-1015 (2013).
  8. Munro, S., Carr, S. M., La Thangue, N. B. Diversity within the pRb pathway: is there a code of conduct. Oncogene. 31 (40), 4343-4352 (2012).
  9. Macdonald, J. I., Dick, F. A. Posttranslational modifications of the retinoblastoma tumor suppressor protein as determinants of function. Genes Cancer. 3 (11-12), 619-633 (2012).
  10. Ezhevsky, S. A., et al. Hypo-phosphorylation of the retinoblastoma protein (pRb) by cyclin D:Cdk4/6 complexes results in active pRb. Proc Natl Acad Sci U S A. 94 (20), 10699-10704 (1997).
  11. Ledl, A., Schmidt, D., Muller, S. Viral oncoproteins E1A and E7 and cellular LxCxE proteins repress SUMO modification of the retinoblastoma tumor suppressor. Oncogene. 24 (23), 3810-3818 (2005).
  12. Li, T., et al. Expression of SUMO-2/3 induced senescence through p53- and pRB-mediated pathways. J Biol Chem. 281 (47), 36221-36227 (2006).
  13. Meng, F., Qian, J., Yue, H., Li, X., Xue, K. SUMOylation of Rb enhances its binding with CDK2 and phosphorylation at early G1 phase. Cell Cycle. 15 (13), 1724-1732 (2016).
  14. Geiss-Friedlander, R., Melchior, F. Concepts in sumoylation: a decade on. Nat Rev Mol Cell Biol. 8 (12), 947-956 (2007).
  15. Flotho, A., Melchior, F. Sumoylation: a regulatory protein modification in health and disease. Annu Rev Biochem. 82, 357-385 (2013).
  16. Eifler, K., Vertegaal, A. C. SUMOylation-Mediated Regulation of Cell Cycle Progression and Cancer. Trends Biochem Sci. 40 (12), 779-793 (2015).
  17. Wilkinson, K. A., Henley, J. M. Mechanisms, regulation and consequences of protein SUMOylation. Biochem J. 428 (2), 133-145 (2010).
  18. Gill, G. SUMO and ubiquitin in the nucleus: different functions, similar mechanisms. Genes Dev. 18 (17), 2046-2059 (2004).
  19. Sharma, P., Kuehn, M. R. SENP1-modulated sumoylation regulates retinoblastoma protein (RB) and Lamin A/C interaction and stabilization. Oncogene. 35 (50), 6429-6438 (2016).
  20. Jakobs, A., et al. Ubc9 fusion-directed SUMOylation (UFDS): a method to analyze function of protein SUMOylation. Nat Methods. 4 (3), 245-250 (2007).
  21. Gareau, J. R., Lima, C. D. The SUMO pathway: emerging mechanisms that shape specificity, conjugation and recognition. Nat Rev Mol Cell Biol. 11 (12), 861-871 (2010).
  22. Bernier-Villamor, V., Sampson, D. A., Matunis, M. J., Lima, C. D. Structural basis for E2-mediated SUMO conjugation revealed by a complex between ubiquitin-conjugating enzyme Ubc9 and RanGAP1. Cell. 108 (3), 345-356 (2002).
  23. Saitoh, H., Hinchey, J. Functional heterogeneity of small ubiquitin-related protein modifiers SUMO-1 versus SUMO-2/3. J Biol Chem. 275 (9), 6252-6258 (2000).
  24. Ayaydin, F., Dasso, M. Distinct in vivo dynamics of vertebrate SUMO paralogues. Mol Biol Cell. 15 (12), 5208-5218 (2004).

Tags

In Vivo Detection Analysis Rb Protein SUMOylation Human Cells Endogenous Conditions Exogenous Conditions Retinoblastoma Proteins Small Ubiquitin-related Modifier (SUMO) Lysine Residues Cellular Processes Protein SUMOylation Complexity Of SUMOylation Detection Method DMEM Penicillin Streptomycin HEK293 Cells Incubation PBS G0 Synchronization Process Growth Medium Cell Cycle Analysis G1 Phase Cells SUMO Assay S-phase Synchronization Double Thymidine Block Method
<em>In Vivo</em> Detection and Analysis of Rb Protein SUMOylation in Human Cells
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Meng, F., Li, X., Ren, H., Qian, J.More

Meng, F., Li, X., Ren, H., Qian, J. In Vivo Detection and Analysis of Rb Protein SUMOylation in Human Cells. J. Vis. Exp. (129), e56096, doi:10.3791/56096 (2017).

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