rePort Report Cell Cycle 11:7, 1-14; April 1, 2012; © 2012 Landes Bioscience This manuscript has been published online, prior to printing. Once the issue is complete and page numbers have been assigned, the citation will change accordingly. The peptidyl prolyl isomerase cyclophilin A localizes at the centrosome and the midbody and is required for cytokinesis John H. Bannon,† Darragh S. O’Donovan,† Susan M.E. Kennelly and Margaret M. Mc Gee* School of Biomolecular and Biomedical Science; Conway Institute of Biomolecular and Biomedical Research; University College Dublin; Belfield, Dublin, Ireland These authors contributed equally to this work. † Key words: cyclophilin A, prolyl isomerase, centrosome, cytokinesis, midbody Abbreviations: CypA, cyclophilin A; Cep55, centrosome protein 55; PPIase, peptidyl prolyl isomerase Failed cytokinesis leads to tetraploidy, which is an important intermediate preceding aneuploidy and the onset of tumorigenesis. The centrosome is required for the completion of cytokinesis through the transport of important components to the midbody; however, the identity of molecular components and the mechanism involved remains poorly understood. In this study, we report that the peptidyl prolyl isomerase cyclophilin A (cypA) is a centrosome protein that undergoes cell cycle-dependent relocation to the midzone and midbody during cytokinesis in Jurkat cells, implicating a role during division. Depletion of cypA does not disrupt mitotic spindle formation or progression through anaphase; however, it leads to cytokinesis defects through an inability to resolve intercellular bridges, culminating in delayed or failed cytokinesis. Defective cytokinesis is also evidenced by an increased prevalence of midbody-arrested cells. Expression of wild-type cypA reverses the cytokinesis defect in knockout cells, whereas an isomerase mutant does not, indicating that the isomerization activity of cypA is required for cytokinesis. In contrast, wild-type cypA and the isomerase mutant localize to the centrosome and midbody, suggesting that localization to these structures is independent of isomerase activity. Depletion of cypA also generates tetraploid cells and supernumerary centrosomes. Finally, colony formation in soft agar is impaired in cypA-knockout cells, suggesting that cypA confers clonogenic advantage on tumor cells. Collectively, this data reveals a novel role for cypA isomerase activity in the completion of cytokinesis and the maintenance of genome stability. © 2012 Landes Bioscience. Do not distribute. Introduction Cytokinesis is the final step in cell division and results in the separation of two daughter cells. It involves the assembly of an actomyosin contractile ring, which constricts the plasma membrane and compacts midzone microtubules to form the midbody, which is located within a thin intercellular bridge that connects two daughter cells.1-3 Membrane vesicles fuse with the ingressing plasma membrane to form a new plasma membrane, and abscission of the intercellular bridge finally separates daughter cells. Failure of cytokinesis leads to tetraploidy and genomic instability, which have been implicated in the onset of tumorigenesis.4-7 The centrosome is a structurally conserved organelle that consists of a pair of centrioles surrounded by pericentriolar material.8 The centrosome is the primary site of microtubule nucleation within the cell, and it orchestrates the formation of the mitotic spindle.9,10 It also anchors a number of regulatory proteins involved in cell cycle progression and DNA replication.11 Centriolin and centrosome protein 55 (cep55) migrate from the centrosome to the midbody and regulate the abscission of the intercellular bridge during cytokinesis through the recruitment of vesicle trafficking and fusion machinery.12,13 While the recruitment of these components is critical for efficient abscission, the biochemical events that regulate the final plasma membrane fusion during abscission are only beginning to emerge. Cyclophilins are members of a class of ubiquitously expressed peptidyl-prolyl isomerases (PPIases), which also include the parvulins, the FK-506 binding proteins and the protein Ser/Thr phosphatase 2A (PP2A) activator PTPA,14-16 which catalyze the cis-trans isomerization of proline amide bonds. Cyclophilins act as chaperone proteins and assist in protein folding;17 however, their true physiological function remains elusive. One family member, cyclophilin A (cypA), interacts with the receptor tyrosine kinase Itk and modulates its activity18 and with the human immunodeficiency virus type 1 capsid protein Gag, increasing viral infectivity.19,20 Additional functions have been attributed to cypA in apoptosis and in chemotaxis, and cypA is the intracellular receptor for the immunosuppressive drug cyclosporine A (CsA).21 *Correspondence to: Margaret M. Mc Gee; Email: [email protected] Submitted: 02/14/12; Accepted: 02/14/12 http://dx.doi.org/10.4161/cc.11.7.19711 www.landesbioscience.com Cell Cycle 1 CypA is overexpressed in hematopoietic malignancies, lung, pancreatic and breast cancer, implicating a role in tumorigenesis;22-25 however, the function of cypA during cell growth remains poorly understood. The parvulin pin1 specifically catalyzes the isomerization of proline residues that are preceded by a phosphorylated serine or threonine26 and regulates cell cycle progression, DNA synthesis, centrosome duplication and cytokinesis, supporting a role in the maintenance of genome stability27-29 and raising interest in prolyl isomerization as a therapeutic target.29 The relationship between prolyl isomerization, malignant transformation and tumor progression is poorly understood; however, it is believed that the normal enzymatic function of PPIases is exploited to promote the growth and survival of tumor cells.30 In support of that, recent evidence reveals that the cistrans interconversion catalyzed by PPIases regulates important signaling events, and isomerization catalyzed by Pin1 is critical for JNK activation in breast cancer cells.31 In this study, we demonstrate a role for cypA in the regulation of mammalian cell division. We show that cypA is a centrosomal protein that forms part of the spindle poles during mitosis and migrates to the midbody during telophase, where it regulates abscission. The isomerase activity of CypA is not required for its targeting to the centrosome or to the midbody; however, it is essential for the timely completion of cytokinesis. Depletion of cypA leads to cytokinesis defects, including the generation of tetraploid cells with elevated centrosome number. Thus, our findings unveil a novel role for phosphorylation-independent isomerization in the regulation of cytokinesis and the maintenance of genome stability, which may have important implications for the initiation and progression of cancer. and Jurkat cells. The absence of GAPDH confirms the absence of contaminating cytosolic proteins (Fig. 1E). Finally, cypA localizes at the centrosome in the absence and presence of nocodazole or taxol, indicating that it does not require an intact microtubule network for localization to the centrosome (Fig. 1F). CypA localizes to the spindle poles during metaphase and early anaphase and migrates to the midzone and midbody during late anaphase and telophase. Endogenous cypA was monitored during the cell cycle by confocal immunofluorescence and was found to form part of the spindle poles during metaphase and early anaphase. During late anaphase, cypA levels are diminished at the spindle poles and become concentrated at the midzone and finally, during telophase cypA becomes concentrated at the midbody connecting two daughter cells (Fig. 2A). The midbody staining of cypA resembled that of the centrosome protein cep55.34 To confirm this, Jurkat cells were co-transfected with GFP-Cep55 and mCherry-α-tubulin to highlight midbody and intercellular bridge formation during cytokinesis (Fig. 2B, top part). Consistent with endogenous cypA and cep55, GFPCypA concentrates at the midbody, whereas the empty vector does not (Fig. 2B, bottom and middle parts). CypA also localizes to the midbody in H1299 and K562 cells (Fig. 2C). Collectively, these results illustrate the cell cycle-dependent relocalization of cypA from the centrosome to the midzone and midbody during mitosis, suggesting potential sites of action during cell division. CypA-deficient cells form a bipolar spindle and proceed through normal anaphase. We investigated a role of cypA in bipolar spindle formation and the progression through anaphase. Wild-type and cypA-/- Jurkat cells were probed with antibodies to detect α-tubulin and pericentrin, which highlight the mitotic spindle and centrosomes, respectively. DNA was highlighted by DAPI staining. No apparent defects in mitotic spindle formation were observed in the two cell lines by confocal immunofluorescence (Fig. 3A). Using phase contrast live cell imaging, the time from cell elongation during anaphase onset to furrow ingression during telophase was measured in single cells, and it was found to be similar in the two cell lines (Fig. 3B). Cells were synchronized in prometaphase using a low dose of nocodazole (0.14 μM) and subsequently released. Western blot analysis of whole-cell extracts isolated at the indicated times after release highlight that the rate of cyclin B and phosphorylated histone H3 degradation during mitotic progression was similar in the two cells lines (Fig. 3C). Cell synchronization was validated by flow cytometry (data not shown). Extracts from unsynchronized cells represent a population predominantly comprised of interphase cells, where low levels of cyclin B and phosphorylated histone H3 were detected (Fig. 3C, lanes 1 and 2). In addition, the spindle assembly checkpoint protein, BubR1 was phosphorylated in the wild-type and cypA-/- cells at 0 h and underwent dephosphorylation, which correlates with cyclin B and phosphorylated histone H3 degradation, suggesting that the spindle assembly checkpoint is satisfied in the two cell lines (Fig. 3C). Finally, BubR1 was detected at the kinetochore in the two cell lines by confocal microscopy following treatment with nocodazole and taxol, which activate the spindle assembly checkpoint (Fig. 3D). Collectively, this data illustrates that the spindle assembly checkpoint is functional in © 2012 Landes Bioscience. Do not distribute. Results CypA is a centrosome protein. We have previously reported a nuclear location for cypA in hematopoietic cells.22 We examined the subcellular localization of cypA more closely and detected a prominent single or paired dot-like structure at the nuclear periphery by high power confocal microscopy that resembled centrosomal structures (Fig. 1A). Cells were double stained with antibodies to detect cypA and the core centrosome protein γ-tubulin, and merged images show co-localization of cypA and γ-tubulin at the centrosome in H1299 lung carcinoma cells, chronic myeloid leukemia cells (K562, KYO.1 and Lama84) and Jurkat T lymphoma cells (Fig. 1A). To confirm the centrosome localization of cypA, a GFP-tagged full-length cypA construct (GFP-CypA) was expressed in Jurkat cells that lack cypA, which was confirmed by western blotting (Fig. 1B), and cells were analyzed by immunofluorescent microscopy. GFP-CypA co-localizes with the centrosome protein centrin, whereas the GFP-empty control vector (GFP-E) does not (Fig. 1C). CypA localizes to a single centrosome in interphase cells and to two centrosomes immediately following S-phase duplication (Fig. 1D). In contrast, centrosomal staining of cypA was not detected in Jurkat cells that stably lack cypA (Fig. 1D). Furthermore, cypA and γ-tubulin co-migrate in enriched centrosome fractions isolated from K562 2 Cell Cycle Volume 11 Issue 7 © 2012 Landes Bioscience. Do not distribute. www.landesbioscience.com Cell Cycle 3 Figure 1 (See previous page). CypA is a centrosome protein. (A) H1299, K562, Lama84, KYO1 and Jurkat cells were prepared on slides and stained with anti-γ-tubulin (green), anti-cypA (red) and DAPI (blue). (B) Jurkat CypA-/- cells were transfected with GFP-CypA or GFP vector control (GFP-E). GFP protein expression was confirmed by western blot with anti-GFP. (C) Jurkat CypA-/- cells expressing GFP-CypA and GFP-E were stained with anti-centrin (red) to visualize centrosomes. (D) Jurkat cells were prepared on slides and stained with anti-γ-tubulin (green), anti-cypA (red) and DAPI (blue). (E) Centrosome extracts were prepared from K562 or Jurkat cells by fractionation on a discontinuous sucrose gradient and resolved using SDS-PAGE followed by western blotting with anti-cypA, anti-γ-tubulin and anti-GAPDH. (F) Jurkat cells were incubated with vehicle (0.5% (v/v) DMSO), 0.1 μM taxol or 0.1 μM nocodazole for 24 hr. Samples were prepared on slides and stained with anti-γ-tubulin (green), anti-cypA (red) and DAPI (blue). Arrowheads indicate co-localization. Bar: 10 μm. wild-type and cypA-/- cells, and the cells proceed from anaphase to telophase with similar kinetics. CypA is required for abscission. The mitotic index of WT and cypA-/- cells was measured using multivariate flow cytometry following treatment with nocodazole and taxol. A representative cell profile illustrates that 2.13% of wild-type cells are in mitosis (contain 4N DNA), and this increased to 9.56% and 17.8% following treatment with nocodazole and taxol, respectively (Fig. 4A, top part). However, while the overall mitotic index is not significantly different in the cypA-/- cells, there was a dramatic shift in the mitotic cell population from 4N to an 8N state. Representative data outlined in Figure 4A shows that 1.87% of vehicle-treated cells contain 8N DNA, which increased to 11.1% and 13.9% following treatment with nocodazole and taxol, respectively, whereas only 2% of mitotic cells contain 4N DNA (Fig. 4A, bottom part). The increase in DNA content of the mitotic population was confirmed in triplicate determinations following treatment with nocodozole and taxol (Fig. 4B). The ability of nocodazole and taxol to effectively block cells in prometaphase confirms our earlier finding that cypA-/- cells have a functional spindle assembly checkpoint. Thus, the polyploidization observed in cypA-/- cells is most likely due to post-anaphase defects in cell division. A role of cypA during cytokinesis was investigated by monitoring the time from intercellular bridge formation during telophase to complete abscission of the bridge in single cells transfected with the midbody marker cep55 (GFP-Cep55) and mCherryα-tubulin to highlight central midbody and intercellular bridge structures, respectively. Representative images from single Jurkat cells are shown in Figure 4C. The time from intercellular bridge formation to final abscission of the bridge is significantly prolonged in cells depleted of cypA. The midbody and intercellular bridge is undetectable in wild-type cells after 60 min, and two daughter cells with a complete plasma membrane are formed within 90 min. However, the midbody and intercellular bridge persist in cypA-/- cells up to 120 min after initial bridge formation, and the cell eventually divided by 150 min (Fig. 4C and Movies S1 and S2). A similar delay was detected in K562 cypA siRNA-transfected cells but was absent from control scramble transfected cells (Movies S3 and S4). The mean time from bridge formation to abscission was 84 min and 134 min for wild-type and cypA-/- Jurkat cells, respectively (Fig. 4D), and 120 min and 280 min for scrambled control and cypA siRNA K562 cells (Fig. 4E). In some instances, the intercellular bridge was not resolved 8 h after telophase onset in the cypA siRNA-knockdown cells. Consistently, a significant increase in Jurkat and K562 midbody-arrested cells was detected 180 min after release from the nocodazole block (Fig. 4F–H) that was not detected at 0 min or 60 min. A similar increase in midbody-arrested cells was detected in an unsynchronized cell population (Fig. 4G). Collectively, this data suggests that loss of cypA expression in a number of cell lines leads to delayed cytokinesis through an inability to resolve the intercellular bridge during abscission. CypA isomerase activity is not required for localization to the centrosome or midbody. We investigated the role of cypA isomerase activity in its localization to the centrosome and midbody. CypA-/- Jurkat cells were transfected with either GFPCep55 as a centrosome and midbody marker, GFP empty vector control (GFP-E), GFP-CypA or the isomerase defective mutant, GFP-R55A. Results illustrate that the GFP-R55A mutant localizes to the centrosome in a manner similar to wild-type cypA and cep55 (Fig. 5A). Furthermore, GFP-R55A concentrated at the midbody during cytokinesis, similar to wild-type GFP-CypA and GFP-Cep55 (Fig. 5B). The staining intensity of GPF-R55A at the centrosome and midbody was similar to wild-type GFPCypA and suggests that the centrosome and midbody localization of cypA in Jurkat cells is not dependent on its isomerase activity. CypA isomerase activity is required for abscission. Cis-trans isomerization is an important regulator of cell cycle progression.35 A role for the isomerization activity of cypA during cytokinesis was investigated through a series of rescue experiments, whereby Jurkat cypA-/- cells that undergo delayed cytokinesis were cotransfected with mCherry-α-tubulin and either GFP vector control (GFP-E), GFP-CypA or the isomerase defective GFP-R55A, and the time from telophase to abscission was measured by live cell imaging as before. The average time to complete cell division was approx 130 min for cypA-/- cells, which is consistent with earlier findings (Fig. 4C). The re-expression of wild-type cypA reduced the time to complete division to approximately 90 min (Fig. 5C) and is similar to that measured in the wild-type Jurkat cells (Fig. 4C). However, transfection of cypA-/- cells with the R55A mutant failed to rescue the division delay observed, and the time taken to complete division did not differ significantly from cypA-/- cells (Fig. 5C). These results outline that the cistrans isomerization catalyzed by cypA in required for the timely completion of cytokinesis. Loss of CypA generates tetraploid cells. Failed cytokinesis is often characterized by bi- or multi-nucleated cells or enlarged mononuclear cells formed through the fusion of bi-nucleates.6 The loss of cypA expression in Jurkat cells led to a decrease in the 2N DNA population with a concomitant increase in 4N and 8N polyploid populations, a phenotype consistent with failed mitosis (Fig. 6A). This was accompanied by a pronounced increase in mononuclear cells with large nuclei (diameter >10 nm) (Fig. 6B and C). The number of bi- and multi-nucleated cells was elevated in © 2012 Landes Bioscience. Do not distribute. 4 Cell Cycle Volume 11 Issue 7 © 2012 Landes Bioscience. Do not distribute. Figure 2. CypA undergoes cell cycle dependent localization to the midzone and midbody. (A) Jurkat cells were prepared on slides and stained with anti-γ-tubulin (green), anti-cypA (red) and DAPI (blue). (B) Jurkat CypA-/- cells were transfected with mCherry-α-tubulin and either GFP-Cep55, GFP vector control (GFP-E) or GFP-CypA. Transfected cells were subjected to videomicroscopy at 30 sec intervals for 3 hr. (C) H1299 and K562 cells stained with anti-γ-tubulin (green) and anti-cypA (red). Arrows highlight midbody. Bar: 10 μm. www.landesbioscience.com Cell Cycle 5 © 2012 Landes Bioscience. Do not distribute. Figure 3. Loss of CypA does not abrogate bipolar spindle formation or progression through mitosis. (A) Jurkat WT and CypA-/- cells were prepared on slides, fixed and stained with anti-pericentrin (red), anti-α-tubulin (green) and DAPI (blue). Representative images from interphase and mitosis are shown. (B) Jurkat cells were observed by phase contrast live cell imaging. The time from cell elongation at anaphase onset to contraction of the cleavage furrow during telophase was recorded, n = 20. (C) Jurkat WT and CypA-/- cells were synchronized in prometaphase. Upon release whole cell lysates were prepared at the indicated times and samples were resolved by SDS-PAGE followed by western blotting with anti-BubR1, anti-cyclin B, anti-Phospho-Histone H3 (PH-H3) and anti-GAPDH. Unsynchronized whole cell extracts were included as a control. (D) Jurkat WT and CypA-/- cells were incubated with vehicle (0.5% (v/v) DMSO), 0.1 μM taxol or 0.1 μM nocodazole for 24 hr. Cells were prepared on slides and stained with anti-BubR1 (green) and DAPI (blue). Error bars are SEM. Bars: 10 μm. 6 Cell Cycle Volume 11 Issue 7 Jurkat and K562 cells following loss of cypA expression (Fig. 6B and D). Increased ploidy was confirmed by metaphase chromosome counts from WT and cypA-/- Jurkat cells, as shown in Figure 6E and F. This data reveals that wild-type cells exhibit a diploid chromosome number, whereas cypA-knockdown cells display a near tetraploid chromosome number. CypA deficiency leads to supernumerary centrosomes and decreased proliferation. Tetraploid cells produced through failed cytokinesis contain multiple centrosomes that lead to multipolar mitosis and eventual aneuploidy.36 Centrosome number was examined in wild-type and cypA-/- Jurkat cells in culture. Early passage cells contained 1 or 2 centrosomes following depletion of cypA; however, a significant increase in centrosome number was detected in late passage cells (P + 20) with a concomitant decrease in the number of cells containing < 2 centrosomes (Fig. 7A). Up to four centrosomes were detected clustered together in mononuclear cells with large nuclei (Fig. 7B). Jurkat and K562 cells deficient in cypA also display a significant reduction in their proliferative capacity compared with wild-type cells in culture (Figs. 7C and S1), which is consistent with the delay in division detected. Finally, the clonogenic potential of wild-type and cypA-/- Jurkat cells was tested in colony-forming assays. Wildtype Jurkat cells display clonogenic ability consistent with previous reports in reference 33, and depletion of cypA significantly reduced colony formation (Fig. 7D), suggesting that overexpression of cypA confers a clonogenic advantage in tumor cells. and progress through normal anaphase.43 In addition, we have shown that the spindle assembly checkpoint is functional and satisfied in the wild-type and cypA-deficient cells, thus enabling anaphase onset, and cells progress from anaphase to telophase with similar kinetics. Furthermore, cypA was not detected at the kinetochore or centromere during anaphase, suggesting that a function in chromosome assembly or segregation during early anaphase is unlikely. Therefore, although cypA is present at the spindle poles, it is dispensable for spindle formation, and cells lacking cypA proceed through an apparently normal anaphase. CypA-deficient cells display a dramatic increase in the proportion of mitotic cells that harbor 8N DNA. These cells have a functional spindle assembly checkpoint and are efficiently arrested in metaphase by microtubule poisons, and the polyploid phenotype observed is consistent with failed division after anaphase.44 Consistent with this, cytokinetic defects were observed in cypA-knockout cells. The predominant phenotype observed is persistence of the intercellular bridge joining two daughter cells for extended periods, and the time to complete division was almost doubled in cypA-knockout cells. This phenotype is similar to that detected following centriolin silencing and following overexpression of the Cdc14A phosphatase.12,45 Imaging data also revealed that the cleavage furrow and intercellular bridge forms with similar kinetics in the two cell lines. Impaired cytokinesis was also shown by an increase in midbody-arrested cells in unsynchronized and synchronized cell populations. Importantly, re-expression of full-length cypA in knockout cells restored the time to complete division to that of wild-type cells; however, in contrast, the isomerase defective mutant did not, and cells continue to display persistent intercellular bridges and prolonged cytokinesis. Collectively, this data reveals that cypA isomerase activity is required for the final stage of cell division and supports the recent finding that isomerization of midbody components is an important regulator of abscission.46 In this study, we also reveal that isomerization activity of cypA is not required for efficient targeting of cypA to the centrosome and midbody. Further research is required to provide insight into the structural basis of cypA at these locations. Failed cytokinesis results in the formation of bi- and multinucleated cells or enlarged mononuclear cells,47 which was detected in Jurkat and K562 cells following loss of cypA expression. Enlarged nuclear volume in Jurkat cells correlates with increased ploidy and is consistent with an increase in 4N and 8N DNA detected and a concomitant decrease in cells with 2N DNA. Cells with > 8N DNA were not detected, suggesting that endoreplication did not occur. Increased ploidy and genomic instability was confirmed in metaphase chromosome spreads, which revealed that cypA deficiency induces tetraploidization and is consistent with that observed during cytokinesis failure following loss of Nek7 kinase activity.47 Genomic instability and tetraploidization is associated with amplified chromosome number.48 Up to four centrosomes were detected in late-passage cypA-deficient Jurkat cells with a concomitant decrease in cells with less than two centrosomes. The proportion of multicentrosomal cells correlates with the number © 2012 Landes Bioscience. Do not distribute. Discussion In recent years, important molecular components of the cytokinetic machinery have been identified that regulate plasma membrane remodeling and membrane severing.37-39 While the recruitment of these components is critical for efficient abscission, the underlying mechanism is not completely understood. Pin1-induced isomerization of cep55 has recently been shown to facilitate its phosphorylation by polo-like kinase 1 (Plk-l) and function during abscission, highlighting the importance of isomerization as a mechanism to control cytokinesis.1-3 In this paper, we report a novel role for phosphorylation-independent cis-trans isomerization during cytokinesis. CypA is localized to the centrosome during interphase in a range of tumor cells, including lung carcinoma, leukemia and lymphoma. Centrosome localization of cypA was confirmed by immunofluorescent imaging of endogenous protein and following the exogenous expression of a full-length GFP-cypA construct and by co-migration with the core centrosome protein γ-tubulin in enriched centrosome fractions. Centrosome localization is independent of a nucleated microtubule array confirming that cypA is a bona fide centrosome component. CypA is present in two centrosomes following S-phase duplication and forms part of the mitotic spindle poles during metaphase. During late anaphase, cypA translocates to the midzone and the midbody, and its localization resembles that of cep5534 and other centrosome proteins, including Plk1, centriolin, PTP-BL and LAPSER-1.12,40-42 Loss of cypA does not abrogate spindle formation in Jurkat cells, which is consistent with reports that acentrosomal cells form mitotic spindles www.landesbioscience.com Cell Cycle 7 © 2012 Landes Bioscience. Do not distribute. Figure 4. (A–C) CypA is required for cytokinesis. Jurkat WT and CypA-/- cells were incubated with vehicle (0.5% (v/v) DMSO), 0.1 μM taxol or 0.1 μM nocodazole for 24 hr and samples were fixed and immunostained with anti-PH-H3 primary antibody followed by AlexaFluor488 secondary antibody and analyzed by flow cytometry. Representative histograms are shown in (A). Boxes indicate mitotic populations. Mean mitotic indices for Jurkat WT and CypA-/- are shown in (B), n = 3. Jurkat WT and CypA-/- cells were transfected with mCherry-α-tubulin and GFP-Cep55 and subjected to videomicroscopy at 30 sec intervals for 3 hr. Representative images up to 150 min are displayed in (C) and the mean time of division is illustrated for Jurkat. of cells with enlarged nuclei. Given that our data reveals a latestage cytokinesis defect, it is most likely that the multi-centrosomal cells arise due to the continued cycling of binucleate cells produced from impaired cytokinesis and is not a consequence of uncontrolled centrosome duplication. In support of this, a large proportion of the tetraploid cells stained positive for the phosphorylated histone H3, confirming that they entered mitosis. Tetraploid cells are inherently unstable and contribute to the tumorigenic process.48 As expected, the centrosome and chromosome abnormalities observed increased after prolonged time in culture. In addition, the proliferative capacity of cypA depleted cells was dramatically impaired after 48 h in culture, which may be a direct result of prolonged or failed cytokinesis. In addition, while it is clear that Jurkat cells can tolerate a certain level of ploidy, some tetraploid cells may undergo multipolar mitosis and subsequent p53-dependent growth arrest and/or cell death which may also contribute to the decrease in cell number detected. 8 Furthermore, the colony-forming ability of cypA-deficient cells was significantly impaired under conditions that supported the colony formation of wild-type cells. This data supports the finding that deregulated cypA confers a growth advantage on tumor cells and is reminiscent of other cytokinesis regulators, including PLK-1 and Aurora B, which confer clonogenic advantage to tumor cells.49 In summary, we have characterized the role for cypA during cell division and have shown that the prolyl isomerization catalyzed by cypA at the midbody is essential for the timely completion of cytokinesis and the maintenance of genomic stability. The midbody acts as a site for the recruitment of secretory vesicles and membrane fusion machinery. Thus, cypA may regulate the functional isomers of important midbody proteins required for abscission. Interestingly, cypA is an important component of the retroviral budding machinery, and it co-localizes with Rab24, implicating a role in vesicular trafficking.50 The identification of Cell Cycle Volume 11 Issue 7 © 2012 Landes Bioscience. Do not distribute. Figure 4. (D–H) CypA is required for cytokinesis. (D) and K562 (E), n = 10. Jurkat WT and CypA-/- cells were unsynchronized or synchronized in prometaphase. At the indicated time after release cells were stained with α-tubulin (green) and DAPI (blue). Representative images from each time point are shown (F) and intercellular bridges were quantified for Jurkat (G) and K562 cells (H) in a field of view of ~50 cells, n = 5. Error bars are SEM *p < 0.05, **p < 0.01. Bars: 10 μm. cypA substrates at the midbody will provide important insight into the molecular mechanism of abscission, which has significant implications for tumorigenesis. Materials and Methods Cell culture and synchronization. Jurkat, KYO1, Lama84, K562 and H-1299 cells were grown in RPMI-1640 medium containing 10% (v/v) FCS, 2 mM L-glutamine and 100 μg/ml penicillin/ www.landesbioscience.com streptomycin solution. Cells were synchronized in mitosis by incubating 0.14 μM nocodazole for 16 hrs and released by washing twice in growth medium. Jurkat CypA-/- cells were obtained through the AIDS Research and Reference Reagent Program and were created as described previously in reference 22. Plasmids and transfections. pmCherry-α-tubulin was created by excising the α-tubulin fragment from pEYFP-Tub (BD Biosciences Clontech) and ligating into pmCherry‑C1 (BD Biosciences Clontech). pEGFP-CypA-WT/R55A plasmids were Cell Cycle 9 © 2012 Landes Bioscience. Do not distribute. Figure 5. Cis-trans isomerization catalyzed by CypA is required for abscission. Jurkat CypA-/- cells were transfected with GFP-Cep55, GFP vector control (GFP-E), GFP-CypA or GFP-R55A. Cells were prepared on slides and were stained with anti-centrin (red). Colocalization is indicated by a yellow color (A). Jurkat CypA-/- cells were transfected with mCherry-α-tubulin and either GFP-Cep55, GFP vector control (GFP-E), GFP-CypA or GFP-R55A. Transfected cells were subjected to videomicroscopy at 30 sec intervals for 3 hr. Representative images are shown (B). Arrows highlight midbody. In (C) expression of GFP-E, GFP-CypA or GFP-R55A was confirmed by western blot using anti-GFP antibody and GAPDH as a loading control. Average time was calculated from telophase to the abscission of the intercellular bridge, n = 10. Error bars are SEM **p < 0.01. Bar: 10 μm. 10 Cell Cycle Volume 11 Issue 7 © 2012 Landes Bioscience. Do not distribute. Figure 6. CypA is required for genome stability. (A) Jurkat WT and CypA-/- cells were fixed, stained with PI and DNA content was quantified by flow cytometry. n = 3. *p < 0.05. Nuclei were evaluated by fluorescent microscopy and the number of nuclei per cell and nuclear size (>10 μm diameter) was tabulated (B). n = 50. *p < 0.05. Jurkat (WT and CypA-/- cells) and K562 (control scrambled and cypA siRNA KD cells) were prepared on slides, stained with DAPI and observed using a Zeiss fluorescent microscope. Representative images of Jurkat WT and CypA-/- cells are shown (C). Multinucleated K562 cells are tabulated (D). Metaphase chromosome spreads from WT and CypA-/- cells were observed using a Zeiss fluorescent microscope. Representative images of WT and CypA-/- cells are shown in (E) and the distribution of chromosomes is shown (F). n = 20. All error bars are SEM. Bar: 10 μm. created by excising CypA-WT/R55A from pcDNA3.1-CypAWT/R55A, a kind gift from Professor Jeremy Luban, Department of Microbiology and Molecular Medicine, University of Geneva, and ligating into or GFP-C1 (BD Biosciences Clontech). pEGFPC3-Cep55 was a kind gift from Professor Kerstin Kutsche, Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf. All sequences were verified by automated sequencing. Plasmid www.landesbioscience.com transfections in Jurkat and K562 cells were performed using the Amaxa biosystems Cell Line Nucleofector® Kit V. Antibodies. The following primary antibodies were used: anti-phospho-histone-H3 (Ser10) (Cell Signaling), anti-cypA and anti-GAPDH (Millipore), anti-γ-tubulin and anti-β-actin (Sigma), anti-BubR1 and anti-cyclin B1 (BD PharMingen), anti-α-tubulin and anti-pericentrin (Abcam), anti-centrin and Cell Cycle 11 © 2012 Landes Bioscience. Do not distribute. Figure 7. Loss of cypA leads to supernumerary centrosomes and decreased proliferation. Jurkat WT and CypA-/- cells were prepared on slides and stained with anti-pericentrin. Centrosome number was counted in early passage (P + 6) and late passage (P + 20) Jurkat cells in a field of 100 cells per experiment (A), n = 3. Representative images are shown (B). (C) WT and Jurkat CypA-/- cells were seeded (5.5 x 106/50 ml) in complete growth medium and cell number was determined after 24, 48, 72 and 96 hr. n = 3. (D) Jurkat WT and CypA-/- cells were seeded in soft agar, incubated for 28 d at 37°C, 95% O2 and 5% CO2 and stained using crystal violet. n = 3. **p < 0.01. anti-GFP (Santa Cruz). Secondary anti-rabbit and anti-mouse HRP conjugated antibodies (Cell Signaling) were used for western blot, and AlexFluor488 or AlexaFluor594 conjugated antirabbit and anti-mouse secondary antibodies (Invitrogen) were 12 used for imaging and flow cytometry. Anti-FLAG antibody was from Sigma-Aldrich. Flow cytometry. Cells were harvested by centrifugation at 450 RCF for 3 min. The cell pellet was resuspended in PBS, and cells were fixed in ice-cold 70% ethanol at 4°C overnight. Cell Cycle Volume 11 Issue 7 Cells were centrifuged at 800 RCF for 5 min and resuspended in PBS containing RNase A (300 μM) and propidium iodide (PI) (200 μM), and samples were incubated in the dark at 37°C for 30 min before analysis. Multivariate analysis was performed using anti-phospho-histone-H3, to label mitotic cells. Cells were fixed in 70% ethanol, blocked in 2% BSA/PBS and incubated with primary antibody. AlexaFluor 488-conjugated secondary antibody (Invitrogen) was used to detect the primary antibody, and samples were co-stained with PI. PI was detected using a 488:613 band pass dichroic, and AlexaFluor488 was detected using a 488:525 band pass dichroic on a CyAn ADP Flow Cytometer. Analysis was performed using Summit v4.3 software. Immunoblotting and immunofluorescence. Immunoblotting was performed as described previously in reference 22. Proteins were visualized by enhanced chemiluminescence solution (Pierce) and X-ray film (Fujifilm). For immunofluorescence, fixation and immunostaining were performed as described previously in reference 32. Cells were incubated with relevant primary antibodies, which were detected with Alexa Fluor 488- or 594-conjugated secondary antibodies. DNA was stained with DAPI (Sigma). Images were acquired at 63x magnification on a Zeiss 510UV Meta confocal microscope at room temperature. Images were captured using the LSM Imaging software. Time-lapse imaging. For phase contrast live cell imaging, Jurkat cells were transferred to multichambered microscopy slides (IBIDI), which were coated with poly-l-lysine (Sigma). Cells were observed using phase contrast at 60x magnification on a Zeiss microscope with an Andor iXonEM EMCCD camera, and images were captured every 30 sec for 2 h. Movies were made using Andor IQ software. For fluorescent imaging, Jurkat cells were transfected with fluorescent plasmids as described and transferred to multichambered microscopy slides (IBIDI), which were coated with poly-l-lysine. Fluorescent proteins were observed using 488 nm and 594 nm laser excitation at 100x magnification on a Nikon confocal spinning disc microscope with an Andor iXonEM EMCCD camera. Images were captured every 30 sec for 3 h at 37°C, 95% O2 and 5% CO2. Movies were made using Andor IQ software. Centrosome preparations. Centrosomes were purified from cells using a sucrose gradient as described previously in reference 33. Ten fractions were collected, and centrosomes were recovered by centrifugation at 15,000 RCF for 10 min, boiled in SDS sample buffer and resolved by SDA-PAGE followed by western blotting. Colony formation assay. A cell suspension (5 x 105 cells/ml) was prepared in a liquid 0.7% agarose/growth media solution, plated onto soft agar as described previously in reference 33 and incubated at 37°C, 95% O2 and 5% CO2. After 28 d, the samples were fixed and stained with 0.2% crystal violet as outlined. Colonies were photographed using a digital camera and counted. Metaphase spread. Cells were incubated with 10 μM nocodazole for 2 hrs at 37°C, 95% O2 and 5% CO2. Cells were centrifuged at 450 RCF for 3 min. The pellet was washed in PBS and resuspended in 900 μl KCl (0.075 μM) and incubated at 37°C for 17 mins. Cells were fixed in 100 μl of cold fixative (3:1, methanol:acetic acid) and centrifuged at 1,500 RCF for 2 mins. The supernatant was removed, the pellet was resuspended, and 20 μl of the sample was dropped onto the center of a glass slide and allowed to dry. The DNA was stained using DAPI. DNA was visualized using a Zeiss fluorescent microscope and Axiovision LE 4.8.1 software. © 2012 Landes Bioscience. Disclosure of Potential Conflicts of Interest Do not distribute. References 1. Barr FA, Gruneberg U. Cytokinesis: placing and making the final cut. Cell 2007; 131:847-60; PMID:18045532; http://dx.doi.org/10.1016/j. cell.2007.11.011. 2. Eggert US, Mitchison TJ, Field CM. Animal cytokinesis: from parts list to mechanisms. Annu Rev Biochem 2006; 75:543-66; PMID:16756502; http://dx.doi. org/10.1146/annurev.biochem.74.082803.133425. 3. Glotzer M. The molecular requirements for cytokinesis. Science 2005; 307:1735-9; PMID:15774750; http:// dx.doi.org/10.1126/science.1096896. 4. Caldwell CM, Green RA, Kaplan KB. APC mutations lead to cytokinetic failures in vitro and tetraploid genotypes in Min mice. J Cell Biol 2007; 178:110920; PMID:17893240; http://dx.doi.org/10.1083/ jcb.200703186. www.landesbioscience.com No potential conflicts of interest were disclosed. Acknowledgements We thank Prof. Dimitri Scholz, Head of the Conway Institute Imaging core facility for help with the microscopy and Dr. Alfonso Blanco for help with flow cytometry. Financial Support This work was supported by Cancer Research Ireland and Science Foundation Ireland. Note Supplemental material can be found at: www.landesbioscience.com/journals/cc/article/19711 5. Fujiwara T, Bandi M, Nitta M, Ivanova EV, Bronson RT, Pellman D. Cytokinesis failure generating tetraploids promotes tumorigenesis in p53-null cells. Nature 2005; 437:1043-7; PMID:16222300; http://dx.doi. org/10.1038/nature04217. 6. Ganem NJ, Storchova Z, Pellman D. Tetraploidy, aneuploidy and cancer. Curr Opin Genet Dev 2007; 17:157-62; PMID:17324569; http://dx.doi. org/10.1016/j.gde.2007.02.011. 7. Steigemann P, Wurzenberger C, Schmitz MH, Held M, Guizetti J, Maar S, et al. Aurora B-mediated abscission checkpoint protects against tetraploidization. Cell 2009; 136:473-84; PMID:19203582; http://dx.doi. org/10.1016/j.cell.2008.12.020. 8. Kalt A, Schliwa M. Molecular components of the centrosome. Trends Cell Biol 1993; 3:118-28; PMID:14731766; http://dx.doi.org/10.1016/09628924(93)90174-Y. 9. Doxsey SJ. Centrosomes as command centres for cellular control. Nat Cell Biol 2001; 3:105-8; PMID:11331889; http://dx.doi.org/10.1038/35074618. Cell Cycle 10. Rieder CL, Faruki S, Khodjakov A. The centrosome in vertebrates: more than a microtubule-organizing center. Trends Cell Biol 2001; 11:413-9; PMID:11567874; http://dx.doi.org/10.1016/S0962-8924(01)02085-2. 11. Doxsey S, McCollum D, Theurkauf W. Centrosomes in cellular regulation. Annu Rev Cell Dev Biol 2005; 21:411-34; PMID:16212501; http://dx.doi. org/10.1146/annurev.cellbio.21.122303.120418. 12. Gromley A, Jurczyk A, Sillibourne J, Halilovic E, Mogensen M, Groisman I, et al. A novel human protein of the maternal centriole is required for the final stages of cytokinesis and entry into S phase. J Cell Biol 2003; 161:535-45; PMID:12732615; http://dx.doi. org/10.1083/jcb.200301105. 13. Martinez-Garay I, Rustom A, Gerdes HH, Kutsche K. The novel centrosomal associated protein CEP55 is present in the spindle midzone and the midbody. Genomics 2006; 87:243-53; PMID:16406728; http:// dx.doi.org/10.1016/j.ygeno.2005.11.006. 13 14. Davis TL, Walker JR, Campagna-Slater V, Finerty PJ, Paramanathan R, Bernstein G, et al. Structural and biochemical characterization of the human cyclophilin family of peptidyl-prolyl isomerases. PLoS Biol 2010; 8:1000439; PMID:20676357; http://dx.doi. org/10.1371/journal.pbio.1000439. 15. Howard BR, Vajdos FF, Li S, Sundquist WI, Hill CP. Structural insights into the catalytic mechanism of cyclophilin A. Nat Struct Biol 2003; 10:475-81; PMID:12730686; http://dx.doi.org/10.1038/nsb927. 16. Wang P, Heitman J. The cyclophilins. Genome Biol 2005; 6:226; PMID:15998457; http://dx.doi. org/10.1186/gb-2005-6-7-226. 17. Göthel SF, Marahiel MA. Peptidyl-prolyl cis-trans isomerases, a superfamily of ubiquitous folding catalysts. Cell Mol Life Sci 1999; 55:423-36; PMID:10228556; http://dx.doi.org/10.1007/s000180050299. 18. Brazin KN, Mallis RJ, Fulton DB, Andreotti AH. Regulation of the tyrosine kinase Itk by the peptidylprolyl isomerase cyclophilin A. Proc Natl Acad Sci USA 2002; 99:1899-904; PMID:11830645; http://dx.doi. org/10.1073/pnas.042529199. 19. Luban J, Bossolt KL, Franke EK, Kalpana GV, Goff SP. Human immunodeficiency virus type 1 Gag protein binds to cyclophilins A and B. Cell 1993; 73:106778; PMID:8513493; http://dx.doi.org/10.1016/00928674(93)90637-6. 20. Sokolskaja E, Sayah DM, Luban J. Target cell cyclophilin A modulates human immunodeficiency virus type 1 infectivity. J Virol 2004; 78:128008; PMID:15542632; http://dx.doi.org/10.1128/ JVI.78.23.12800-8.2004. 21. Schreiber SL, Crabtree GR. The mechanism of action of cyclosporin A and FK506. Immunol Today 1992; 13:136-42; PMID:1374612; http://dx.doi. org/10.1016/0167-5699(92)90111-J. 22. Bane FT, Bannon JH, Pennington SR, Campiani G, Williams DC, Zisterer DM, et al. The microtubuletargeting agents, PBOX-6 [pyrrolobenzoxazepine7-[(dimethylcarbamoyl)oxy]-6-(2-naphthyl)pyrrolo[2,1-d](1,5)-benzoxazepine] and paclitaxel, induce nucleocytoplasmic redistribution of the peptidyl-prolyl isomerases, cyclophilin A and pin1, in malignant hematopoietic cells. J Pharmacol Exp Ther 2009; 329:3847; PMID:19131583; http://dx.doi.org/10.1124/ jpet.108.148130. 23. Howard BA, Furumai R, Campa MJ, Rabbani ZN, Vujaskovic Z, Wang XF, et al. Stable RNA interferencemediated suppression of cyclophilin A diminishes non-small-cell lung tumor growth in vivo. Cancer Res 2005; 65:8853-60; PMID:16204056; http://dx.doi. org/10.1158/0008-5472.CAN-05-1219. 24. Li M, Zhai Q, Bharadwaj U, Wang H, Li F, Fisher WE, et al. Cyclophilin A is overexpressed in human pancreatic cancer cells and stimulates cell proliferation through CD147. Cancer 2006; 106:2284-94; PMID:16604531; http://dx.doi.org/10.1002/ cncr.21862. 25. Zheng J, Koblinski JE, Dutson LV, Feeney YB, Clevenger CV. Prolyl isomerase cyclophilin A regulation of Janus-activated kinase 2 and the progression of human breast cancer. Cancer Res 2008; 68:7769-78; PMID:18829531; http://dx.doi.org/10.1158/00085472.CAN-08-0639. 26. Ranganathan R, Lu KP, Hunter T, Noel JP. Structural and functional analysis of the mitotic rotamase Pin1 suggests substrate recognition is phosphorylation dependent. Cell 1997; 89:875-86; PMID:9200606; http://dx.doi.org/10.1016/S0092-8674(00)80273-1. 27. Suizu F, Ryo A, Wulf G, Lim J, Lu KP. Pin1 regulates centrosome duplication, and its overexpression induces centrosome amplification, chromosome instability and oncogenesis. Mol Cell Biol 2006; 26:146379; PMID:16449657; http://dx.doi.org/10.1128/ MCB.26.4.1463-79.2006. 28. Wulf GM, Ryo A, Wulf GG, Lee SW, Niu T, Petkova V, et al. Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcriptional activity of c-Jun towards cyclin D1. EMBO J 2001; 20:3459-72; PMID:11432833; http://dx.doi. org/10.1093/emboj/20.13.3459. 29. Yeh ES, Means AR. PIN1, the cell cycle and cancer. Nat Rev Cancer 2007; 7:381-8; PMID:17410202; http:// dx.doi.org/10.1038/nrc2107. 30. Theuerkorn M, Fischer G, Schiene-Fischer C. Prolyl cis/trans isomerase signalling pathways in cancer. Curr Opin Pharmacol 2011; 11:281-7; PMID:21497135; http://dx.doi.org/10.1016/j.coph.2011.03.007. 31. Park JE, Lee JA, Park SG, Lee DH, Kim SJ, Kim HJ, et al. A critical step for JNK activation: isomerization by the prolyl isomerase Pin1. Cell Death Differ 2011; PMID:21660049. 32. Bornens M, Paintrand M, Berges J, Marty MC, Karsenti E. Structural and chemical characterization of isolated centrosomes. Cell Motil Cytoskeleton 1987; 8:238-49; PMID:3690689; http://dx.doi.org/10.1002/ cm.970080305. 33. Hsu CP, Yang CC, Yang SD. Suppression of prolinedirected protein kinase F(A) systemically inhibits the growth of human acute leukemia cells. Int J Cancer 2001; 91:650-3; PMID:11267978; http://dx.doi. org/10.1002/1097-0215(200002)9999:9999<::AIDIJC1100>3.0.CO;2-3. 34. Zhao WM, Seki A, Fang G. Cep55, a microtubulebundling protein, associates with centralspindlin to control the midbody integrity and cell abscission during cytokinesis. Mol Biol Cell 2006; 17:388196; PMID:16790497; http://dx.doi.org/10.1091/mbc. E06-01-0015. 35. Lu KP, Zhou XZ. The prolyl isomerase PIN1: a pivotal new twist in phosphorylation signalling and disease. Nat Rev Mol Cell Biol 2007; 8:904-16; PMID:17878917; http://dx.doi.org/10.1038/nrm2261. 36. Margolis RL. Tetraploidy and tumor development. Cancer Cell 2005; 8:353-4; PMID:16286243; http:// dx.doi.org/10.1016/j.ccr.2005.10.017. 37. Connell JW, Lindon C, Luzio JP, Reid E. Spastin couples microtubule severing to membrane traffic in completion of cytokinesis and secretion. Traffic 2009; 10:42-56; PMID:19000169; http://dx.doi. org/10.1111/j.1600-0854.2008.00847.x. 38. Yang D, Rismanchi N, Renvoisé B, LippincottSchwartz J, Blackstone C, Hurley JH. Structural basis for midbody targeting of spastin by the ESCRT-III protein CHMP1B. Nat Struct Mol Biol 2008; 15:127886; PMID:18997780; http://dx.doi.org/10.1038/ nsmb.1512. 39. Sagona AP, Nezis IP, Pedersen NM, Liestøl K, Poulton J, Rusten TE, et al. PtdIns(3)P controls cytokinesis through KIF13A-mediated recruitment of FYVECENT to the midbody. Nat Cell Biol 2010; 12:36271; PMID:20208530; http://dx.doi.org/10.1038/ ncb2036. 40. Barr FA, Silljé HH, Nigg EA. Polo-like kinases and the orchestration of cell division. Nat Rev Mol Cell Biol 2004; 5:429-40; PMID:15173822; http://dx.doi. org/10.1038/nrm1401. 41. Herrmann L, Dittmar T, Erdmann KS. The protein tyrosine phosphatase PTP-BL associates with the midbody and is involved in the regulation of cytokinesis. Mol Biol Cell 2003; 14:230-40; PMID:12529439; http://dx.doi.org/10.1091/mbc.E02-04-0191. 42. Sudo H, Maru Y. LAPSER1 is a putative cytokinetic tumor suppressor that shows the same centrosome and midbody subcellular localization pattern as p80 katanin. FASEB J 2007; 21:2086-100; PMID:17351128; http://dx.doi.org/10.1096/fj.067254com. 43. Khodjakov A, Cole RW, Oakley BR, Rieder CL. Centrosome-independent mitotic spindle formation in vertebrates. Curr Biol 2000; 10:59-67; PMID:10662665; http://dx.doi.org/10.1016/S09609822(99)00276-6. 44. Meng X, Fan J, Shen Z. Roles of BCCIP in chromosome stability and cytokinesis. Oncogene 2007; 26:6253-60; PMID:17452982; http://dx.doi. org/10.1038/sj.onc.1210460. 45. Kaiser BK, Zimmerman ZA, Charbonneau H, Jackson PK. Disruption of centrosome structure, chromosome segregation and cytokinesis by misexpression of human Cdc14A phosphatase. Mol Biol Cell 2002; 13:2289300; PMID:12134069; http://dx.doi.org/10.1091/ mbc.01-11-0535. 46. van der Horst A, Khanna KK. The peptidyl-prolyl isomerase Pin1 regulates cytokinesis through Cep55. Cancer Res 2009; 69:6651-9; PMID:19638580; http://dx.doi.org/10.1158/0008-5472.CAN-09-0825. 47. Salem H, Rachmin I, Yissachar N, Cohen S, Amiel A, Haffner R, et al. Nek7 kinase targeting leads to early mortality, cytokinesis disturbance and polyploidy. Oncogene 2010; 29:4046-57; PMID:20473324; http://dx.doi.org/10.1038/onc.2010.162. 48. Storchova Z, Pellman D. From polyploidy to aneuploidy, genome instability and cancer. Nat Rev Mol Cell Biol 2004; 5:45-54; PMID:14708009; http:// dx.doi.org/10.1038/nrm1276. 49. de Cárcer G, Pérez de Castro I, Malumbres M. Targeting cell cycle kinases for cancer therapy. Curr Med Chem 2007; 14:969-85; PMID:17439397; http://dx.doi.org/10.2174/092986707780362925. 50. Wu M, Yin G, Zhao X, Ji C, Gu S, Tang R, et al. Human RAB24, interestingly and predominantly distributed in the nuclei of COS-7 cells, is colocalized with cyclophilin A and GABARAP. Int J Mol Med 2006; 17:749-54; PMID:16596256. © 2012 Landes Bioscience. Do not distribute. 14 Cell Cycle Volume 11 Issue 7
© Copyright 2025 Paperzz