Fanconi anemia (FA) is an autosomal recessive disorder characterized by developmental defects, bone marrow failure, and cancer susceptibility. Cells derived from FA patients are sensitive to crosslinking agents and have a prolonged G2 phase, suggesting a cell cycle abnormality. Although transfection of type-C FA cells with the FAC cDNA corrects these cellular abnormalities, the molecular function of the FAC polypeptide remains unknown. In the current study we show that expression of the FAC polypeptide is regulated during cell cycle progression. In synchronized HeLa cells, FAC protein expression increased during S phase, was maximal at the G2 /M transition, and declined during M phase. In addition, the FAC protein coimmunoprecipitated with the cyclin-dependent kinase, cdc2. We next tested various mutant forms of the FAC polypeptide for binding to cdc2. A patient-derived mutant FAC polypeptide, containing a point mutation at L554P, failed to bind to cdc2. The FAC/cdc2 binding interaction therefore correlated with the functional activity of the FAC protein. Moreover, binding of FAC to cdc2 was mediated by the carboxyl-terminal 50 amino acids of FAC in a region of the protein required for FAC function. Taken together, our results suggest that the binding of FAC and cdc2 is required for normal G2 /M progression in mammalian cells. Absence of a functional interaction between FAC and cdc2 in FA cells may underlie the cell cycle abnormality and clinical abnormalities of FA.

FANCONI ANEMIA (FA) is an autosomal recessive disease characterized by congenital abnormalities, aplastic anemia, and cancer susceptibility.1-3 Compared with normal cells, FA cells exhibit genomic instability, increased percentage of cells in the G2 phase of the cell cycle, and increased cellular sensitivity to crosslinking agents.2,4,5 Somatic cell hybrid studies have revealed at least five complementation groups of FA.6-9 The cDNAs for two complementation groups (types A and C) have been cloned,10-13 and the chromosomal locus of one additional FA gene (type D) has been mapped.14 Cells derived from mice with targeted disruptions of the FAC gene also exhibit genomic instability comparable with that observed in FA cell lines.15,16 Although the function of the FAC polypeptide remains unknown, the protein has been reported to be localized to the cytoplasm and is therefore unlikely to play a direct role in DNA repair.17,18 

Several studies suggest that FA results from a molecular defect in cell cycle progression. First, FA cells have a prolonged G2 transit time,19 which is enhanced by treatment with chemical crosslinking agents.20 Second, the G2 arrest and reduced proliferation of FA cells can be partially corrected by overexpression of a protein, SPHAR, which is a member of the cyclin family of proteins.21 Third, caffeine abrogates the G2 arrest of FA cells.22,23 Consistent with these results, caffeine is known to constitutively activate cdc224 and to override a normal G2 cell cycle checkpoint, which may be defective in FA cells. Fourth, other diseases characterized by genomic instability, such as Li-Fraumeni25 and ataxia telangiectasia (AT),26 result from abnormalities in cell cycle regulation.

The cell cycle abnormality, as evidenced by excessive percentage of G2 , and genomic instability of FA are independent of the p53 pathway. p53 is a tumor suppressor protein known to play an important role in G1 /S25,27 and in G2 /M transitions.28 Unlike AT cells,26 FA cells show normal p53 induction in response to MMC and other DNA damaging agents.29,30 Moreover, once p53 is induced in FA cells p53 dependent apoptosis is normal. Also, following treatment with MMC, FA cells and normal cells exhibit no differences in cell cycle progression from G1 phase to S phase. Taken together, these results suggest that p53 induction and G1 /S transition is normal in FA cells.

The regulation of cell cycle progression from G2 to M phase in mammalian cells has become better described. cdc2 is a cyclin-dependent kinase that assembles with cyclin A and cyclin B and directs G2 progression.31,32 Activated cdc2 phosphorylates multiple substrates, including p53, histone H1, and laminin, and regulates G2 progression. cdc2 is regulated posttranslationally in a series of phosphorylation and dephosphorylation events.33-35 The phosphorylation of cdc2 by the kinase wee1 results in decreased cdc2 kinase activity. The dephosphorylation of cdc2 by the phosphatase cdc25 results in increased cdc kinase activity and progression of the cell cycle from G2 to M phase. Interestingly, the control of activation of cdc2 by wee1 and cdc25 is sensitive to DNA damage.36-41 Cellular treatment with cisplatin, for example, results in increased tyrosine phosphorylation of cdc2 and cell cycle arrest at G2 /M.

In the current study, we show that expression of the FAC protein is regulated during the cell cycle, with peak levels observed at the G2 /M transition period. Also, FAC forms a physical complex with cdc2. Interestingly, FAC interacts with cdc2 through the carboxyl terminal 50 amino acids of FAC, known to be required for its biological activity. We hypothesize that this physical interaction between FAC and cdc2 may underlie the cell cycle abnormality of FA cells.

Cell culture.Epstein-Barr virus (EBV) transformed lymphoblasts were maintained in suspension cultures in RPMI 1640 media supplemented with 15% heat-inactivated fetal calf serum (FCS) and grown in a humidified 5% CO2 -containing atmosphere at 37°C.10,17 A type-C FA lymphoblast cell line, HSC536N, was transfected with the wild-type FAC cDNA, as previously described.10,17 The resulting corrected cell line, HSC536N (+FAC), was maintained in the same conditions with the addition of 100 μg/mL hygromycin. HeLa cells were maintained in DMEM media and 10% FCS and grown in plates at 37°C. The NSK cell line, an EBV-immortalized line made from normal adult peripheral blood (PB) lymphocytes, was obtained from American Type Culture Collection (Rockville, MD).

Fig. 1.

The expression of FAC is regulated during cell cycle progression. HeLa cells were synchronized at G1 /S by double thymidine block and released into regular media. Cell aliquots (1 mL) from the indicated timepoints were washed, suspended in propidium iodide, and assayed by FACS analysis, as described in Materials and Methods. The percentage of cells in each phase of the cell cycle was determined by analyzing FACS data with the computer program CELLFIT (Becton Dickinson). Synchronized cells from each time interval were lysed, whole cell extracts were prepared, and cellular proteins were immunoprecipitated and immunoblotted with an antiserum against FAC (top) or immunoprecipitated with monoclonal against cdc2 followed by immunoblotting with anti–phosphotyrosine-cdc2 (middle) or anti-cdc2 (bottom). Immunoblots were scanned and analyzed by NIH Image. Values along the y-axis are in relative density units and are plotted against time in hours after release from G1 /S synchrony. Each plot is placed along side the corresponding immunoblot with symbols as follows: FAC (•); tyrosine phosphorylated cdc2 (▵); and total cdc2 (□).

Fig. 1.

The expression of FAC is regulated during cell cycle progression. HeLa cells were synchronized at G1 /S by double thymidine block and released into regular media. Cell aliquots (1 mL) from the indicated timepoints were washed, suspended in propidium iodide, and assayed by FACS analysis, as described in Materials and Methods. The percentage of cells in each phase of the cell cycle was determined by analyzing FACS data with the computer program CELLFIT (Becton Dickinson). Synchronized cells from each time interval were lysed, whole cell extracts were prepared, and cellular proteins were immunoprecipitated and immunoblotted with an antiserum against FAC (top) or immunoprecipitated with monoclonal against cdc2 followed by immunoblotting with anti–phosphotyrosine-cdc2 (middle) or anti-cdc2 (bottom). Immunoblots were scanned and analyzed by NIH Image. Values along the y-axis are in relative density units and are plotted against time in hours after release from G1 /S synchrony. Each plot is placed along side the corresponding immunoblot with symbols as follows: FAC (•); tyrosine phosphorylated cdc2 (▵); and total cdc2 (□).

Close modal

Cell synchronization.HeLa cells were synchronized as previously described.42 Briefly, cells (5 × 105/mL) were treated sequentially with 2 mmol/L thymidine for 17 hours, thymidine-free media for 10 hours, and an additional 2 mmol/L thymidine for 12 hours to arrest the cell cycle at the G1 /S boundary. The cells were then released into growth media (RPMI 1640 + 15% FCS) and analyzed at various time intervals. We have performed this method multiple times with consistent cell cycle kinetics. Nocodazole (2 μmol/L) was added for 8 hours after release to enrich for cell populations in G2 /M phase. Synchronized cells were analyzed at different timepoints by staining with propidium iodide as previously described.43 Approximately 10,000 cells were analyzed, and flow histograms were generated (Becton Dickinson, Franklin Lakes, NJ).

Mitomycin C treatment.Asynchronous cells or cells synchronized at G1 /S were incubated with MMC (0.1 μmol/L) for 24 hours. Asynchronous cells were treated for 24 hours and analyzed before the onset of apoptosis.30 Cells were obtained (5 × 105), washed in phosphate-buffered saline (PBS), and prepared for FACS analysis as above.

Metabolic labeling and immunoprecipitation.Metabolic labeling of cells with 35S-methionine and immunoprecipitation (IP) was performed as described previously.44 Whole cell extracts or extracts fractionated from nucleus or cytoplasm were prepared as previously described.17 Extracts (1-mL vol) in TBS (50 mmol/L Tris, 150 mmol/L NaCl, 1% Triton X-100, 0.2% sodium dodecyl sulfate [SDS]) were precleared with preimmune serum and mixed with an affinity purified anti-FAC antiserum (2 μg).17 Protein A–sepharose bound immune complexes were washed three times in TBS containing 1% Triton X-100 and 0.1% SDS and subjected to 10% polyacrylamide SDS gel electrophoresis (PAGE). Following electrophoresis, gels were sequentially washed in concentrated acetic acid, 2,5-diphenyloxazole (PPO), and glycerol. Dried gels were exposed to XAR50 film (Kodak, Rochester, NY) at −70°C.

Absorption of cellular proteins with glutathione-S-transferase fusion proteins.Glutathione-S-transferase fusion proteins containing the amino terminal 100 amino acids of FAC (GST-N), the carboxyl terminal 280 amino acids of FAC (GST-FAC), the carboxyl terminal 280 amino acids of FAC containing the L554P mutation (GST-L554P), and the carboxyl terminal 50 amino acids of FAC (GST-C1) were cloned into the pGEX2T vector (Clontech, Palo Alto, CA), as previously described.17 GST-fusion proteins were prepared by expression in Escherichia coli, cell lysis, incubation with glutathione-sepharose beads, and elution with glutathione. Protein concentrations were determined by the method of Bradford.45 GST-fusion protein (5 μg) was prebound to glutathione sepharose beads (Pharmacia, Piscataway, NJ; 50 μL) in 250 μL of lysis buffer. Washed beads were then added to cell lysates (2 mg protein/mL) prepared from HeLa cells. Lysates were precleared with an overnight incubation with GST bound to glutathione-sepharose beads. The beads were washed three times (0.1% Triton-X 100, 50 mmol/L Tris pH 7.4, 150 mmol/L NaCl) and boiled in loading buffer (100 mmol/L sodium phosphate pH 7.2, 1% SDS, 10% glycerol, 0.001% bromophenol blue) before SDS-PAGE.

Western blotting.Proteins electrophoresed on SDS-PAGE gels were transferred to nitrocellulose in transfer buffer (Tris 25 mmol/L, glycine 200 mmol/L) at 400 mA at 4°C. Ponceau stain (Sigma) was performed to ensure equal loading. Filters were blocked for 1 hour in 5% milk in TBS (50 mmol/L Tris-HCl, pH 8.0, 150 mmol/L NaCl) and were incubated in primary antibody in TBS plus Tween20 (TBS-T) overnight. After wash in TBS-T and exposure to horseradish peroxidase-protein A, enzyme-linked chemiluminescence (Amersham, Arlington Heights, IL) was performed. Densitometry was performed on Western blots by scanning into a computer file, followed by analysis using NIH Image.

Expression of the FAC polypeptide is regulated during the cell cycle.Initially, we analyzed the expression of FAC during the cell cycle. HeLa cells were synchronized by double thymidine block,42 released into S phase, and analyzed at different times (Fig 1). FAC protein was immunoprecipitated and immunoblotted with an anti-FAC antiserum (Fig 1, FAC immunoblot). FAC expression increased during S phase (hours 4 to 6), reached a maximum at G2 /M (hour 7), and declined during mitosis (hour 9). To ensure synchrony throughout the cell cycle, cdc2 was immunoprecipitated and immunoblotted from the same cell extracts. Previous studies have shown that cdc2 remains tyrosine phosphorylated until the G2 /M transition when it is dephosphorylated by cdc25.32,33 Cdc2 was dephosphorylated at 9 hours following release from double thymidine block (Fig 1, phospho-cdc2 immunoblot), showing cell synchronization throughout S phase and G2 phase. Unphosphorylated cdc2 protein levels remained constant throughout the cell cycle, whereas phosphorylated forms increased and then decreased (Fig 1, cdc2 immunoblot), consistent with previous studies.32,46 To quantify the protein levels detected by the immunoprecipitation/Western blot experiment, we analyzed the autoradiography by densitometry (NIH Image). The densitometry quantification, expressed in relative density units, confirmed the increase and decrease in the amount of FAC protein and tyrosine phosphorylated cdc2 during the cell cycle, whereas total cdc2 remained relatively stable.

Fig. 2.

FAC localization does not change during the cell cycle. (A) HeLa cells were synchronized at G1 /S by double thymidine block and released into growth media. Cell synchrony was analyzed by FACS, as described in Materials and Methods. The percentage of cells in each phase of the cell cycle was determined by analyzing FACS data with the computer program CELLFIT (Becton Dickinson).

(B) Cells were metabolically labeled with 35S-methionine during the 2-hour intervals indicated. Proteins from whole cells (T), nuclear (N) extracts, or cytoplasmic (C) extracts were immunoprecipitated with anti-FAC antibody. Alternatively, protein from whole cells was immunoprecipitated with a preimmune serum (P). Immune complexes were resolved by SDS-PAGE. Molecular weight markers are in kilodaltons.

Fig. 2.

FAC localization does not change during the cell cycle. (A) HeLa cells were synchronized at G1 /S by double thymidine block and released into growth media. Cell synchrony was analyzed by FACS, as described in Materials and Methods. The percentage of cells in each phase of the cell cycle was determined by analyzing FACS data with the computer program CELLFIT (Becton Dickinson).

(B) Cells were metabolically labeled with 35S-methionine during the 2-hour intervals indicated. Proteins from whole cells (T), nuclear (N) extracts, or cytoplasmic (C) extracts were immunoprecipitated with anti-FAC antibody. Alternatively, protein from whole cells was immunoprecipitated with a preimmune serum (P). Immune complexes were resolved by SDS-PAGE. Molecular weight markers are in kilodaltons.

Close modal

We next examined the cellular localization of the FAC polypeptide during the cell cycle (Fig 2). In a separate experiment, HeLa cells were again synchronized at G1 /S, using a double thymidine block (Fig 2A). Following release from the block, the progression of the cells through the cell cycle was monitored by DNA flow histograms every 2 hours. During each 2-hour period shown, cells were metabolically labeled with 35S-methionine, lysed, and fractionated into cytoplasmic and nuclear fractions.17 Labeled proteins were immunoprecipitated with an anti-FAC antiserum. The FAC polypeptide was localized to the cytoplasmic fraction throughout the cell cycle (compare lanes 3, 7, 12, 15, and 19). Although metabolic labeling and IP is not as quantitative as IP followed by Western blot, this experiment suggests that FAC expression was maximal during the period 6 to 8 hours after release from G1 /S arrest (lane 14), consistent with results in Fig 1. A 34-kD protein coimmunoprecipitated with FAC from total cell lysates prepared from cells labeled 4 to 6 hours (lane 10) and 6 to 8 hours (lane 14) after release.

cdc2 binds to the FAC polypeptide.We next used a coimmunoprecipitation procedure to identify proteins that bind to the FAC polypeptide (Fig 3). Given the abnormal cell cycle phenotype of FA cells and the presence of a 34-kD protein (p34) in Fig 2, we tested if p34 was immunoreactive with a specific monoclonal antibody against a cyclin-dependent kinase, cdc2. An immunoprecipitation with FAC antiserum followed by immunoblotting with a monoclonal antibody to cdc2 (Santa Cruz, Santa Cruz, CA) identified p34 as a cdc2 (lane 2).

Fig. 3.

The FAC polypeptide binds to the cyclin-dependent kinase, cdc2. Proteins from HSC536N (+FAC) were immunoprecipitated with a preimmune (lane 1) or with anti-FAC affinity purified antibody (lane 2). SDS-PAGE gel was blotted onto nitrocellulose and probed with anti-cdc2 monoclonal antibody or anti-FAC antibody. Molecular weight markers are in kilodaltons.

Fig. 3.

The FAC polypeptide binds to the cyclin-dependent kinase, cdc2. Proteins from HSC536N (+FAC) were immunoprecipitated with a preimmune (lane 1) or with anti-FAC affinity purified antibody (lane 2). SDS-PAGE gel was blotted onto nitrocellulose and probed with anti-cdc2 monoclonal antibody or anti-FAC antibody. Molecular weight markers are in kilodaltons.

Close modal

cdc2 binding correlates with the functional activity of FAC.Previous studies have shown that expression of wild-type FAC polypeptide in FA(C) cells confers MMC resistance and corrects the G2 abnormality of FA(C) lymphoblasts.10,43 An FA(C) cell line, HSC536N (mock), expressing a mutant form of the FAC polypeptide, FAC(L554P), accumulated in the G2 phase following treatment with MMC, 0.1 μmol/L, for 24 hours (Fig 4A, upper right). An isogenic cell line, HSC536N (+FAC), transfected with the wild-type FAC cDNA, displayed less accumulation in G2 phase (Fig 4A, lower right). We next tested for the differential ability of wild-type FAC protein and mutant FAC protein to bind to cdc2 (Fig 4B). The mutant FAC protein (FAC L554P) was immunoprecipitated from HSC536N (mock) cells by two independent anti-FAC antisera (Fig 4B, FAC immunoblot, lanes 2 and 4). The mutant FAC protein had the same electrophoretic mobility as the wild-type FAC protein immunoprecipitated from the HSC536N (+FAC) cells (FAC immunoblot, lanes 1 and 3). Interestingly, the wild-type FAC protein coimmunoprecipitated with cdc2 (Fig 4B, cdc2 immunoblot, lanes 1 and 3). Mutant FAC protein (L554P) failed to coimmunoprecipitate with cdc2 (cdc2 immunoblot, lanes 2 and 4).

Fig. 4.

The FAC/cdc2 interaction correlates with the functional activity of FAC. (A) HSC536N (mock) cells and HSC536N (+FAC) cells were harvested following MMC treatment, stained with propidium iodide, and analyzed by FACS, as described in Materials and Methods. (B) Whole cell extracts from HSC536N (mock) cells (lanes 2 and 4) or HSC536N (+FAC) cells (lanes 1 and 3) were immunoprecipitated with affinity purified anti-FAC antisera prepared from two different New Zealand white rabbits. Immunoprecipitated proteins were electrophoresed by SDS-PAGE, blotted to nitrocellulose, and probed with anti-cdc2 antiserum or anti-FAC antiserum. Molecular weight markers are in kilodaltons.

(C) Whole cell extracts from HSC536N (mock) cells (lane 1) or HSC536N (+FAC) cells (lane 2) were immunoprecipitated with anti-cdc2 monoclonal antibody. Immunoprecipitated proteins were electrophoresed by SDS-PAGE, blotted to nitrocellulose, and probed with affinity purified anti-FAC antisera. Molecular weight markers in kilodaltons.

Fig. 4.

The FAC/cdc2 interaction correlates with the functional activity of FAC. (A) HSC536N (mock) cells and HSC536N (+FAC) cells were harvested following MMC treatment, stained with propidium iodide, and analyzed by FACS, as described in Materials and Methods. (B) Whole cell extracts from HSC536N (mock) cells (lanes 2 and 4) or HSC536N (+FAC) cells (lanes 1 and 3) were immunoprecipitated with affinity purified anti-FAC antisera prepared from two different New Zealand white rabbits. Immunoprecipitated proteins were electrophoresed by SDS-PAGE, blotted to nitrocellulose, and probed with anti-cdc2 antiserum or anti-FAC antiserum. Molecular weight markers are in kilodaltons.

(C) Whole cell extracts from HSC536N (mock) cells (lane 1) or HSC536N (+FAC) cells (lane 2) were immunoprecipitated with anti-cdc2 monoclonal antibody. Immunoprecipitated proteins were electrophoresed by SDS-PAGE, blotted to nitrocellulose, and probed with affinity purified anti-FAC antisera. Molecular weight markers in kilodaltons.

Close modal

To further confirm the binding of cdc2 with wild-type FAC, we next performed a reciprocal IP (Fig 4C). cdc2 was immunoprecipitated from either HSC536N (mock) or HSC536N (+FAC) cells. Again, wild-type FAC and a naturally occurring truncated variant of FAC, Fanconi-related protein 50 (FRP-50),17 coimmunoprecipitated with cdc2 (FAC immunoblot, lane 2). Mutant FAC failed to bind cdc2 (lane 1).

These data suggest that mutant FAC fails to bind directly to cdc2 or fails to colocalize to the proper cellular compartment in which cdc2 binding occurs. The cdc2 binding activity of the FAC protein therefore correlates with its ability to correct the G2 cell cycle abnormality.

cdc2 specifically binds to the carboxyl terminus of FAC.Previous studies have shown that the carboxyl terminus of the FAC protein is required for its biological activity.10,43,47-49 To assess a direct binding interaction between FAC and cdc2, we prepared GST fusion proteins containing variable regions of the FAC polypeptide (Fig 5A). The GST-FAC fusion proteins were prebound to glutathione sepharose and incubated with cellular proteins prepared from HeLa cells first synchronized by double thymidine block at G1 /S and then released for 8 hours in the presence of nocodazole to enrich for cells at G2 /M. All fusion proteins were added at comparable levels, as judged by anti-GST immunoblot (Fig 5B, GST immunoblot). Fusion proteins containing the wild-type carboxyl terminus of FAC bound to cdc2 (Fig 5B, cdc2 immunoblot, lanes 3 and 5), whereas fusion proteins derived from the N-terminus of FAC (lane 2) or containing the carboxyl terminus with the L554P missense mutation (lane 4) failed to bind to cdc2. Taken together, these results suggest that the binding of FAC and cdc2 is mediated by the carboxyl terminal 50 amino acids of FAC.

Fig. 5.

cdc2 specifically binds to the carboxyl terminus of FAC. (A) GST fusion proteins used for in vitro mixing experiments are shown schematically. (B) Whole cell extract (unlabeled) was made from HeLa cells first synchronized at G1 /S by double thymidine block and then released for 8 hours in nocodazole.41 GST (lane 1), GST-N (lane 2), GST-FAC (wild-type; lane 3), GST-FAC (L554P, lane 4), or GST-C1 (lane 5), prebound to glutathione-sepharose beads, was mixed with the extract. Bound cellular proteins were electrophoresed by SDS-PAGE and transferred to nitrocellulose. The filter was probed with either anti-GST monoclonal antibody or anti-cdc2 monoclonal antibody.

Fig. 5.

cdc2 specifically binds to the carboxyl terminus of FAC. (A) GST fusion proteins used for in vitro mixing experiments are shown schematically. (B) Whole cell extract (unlabeled) was made from HeLa cells first synchronized at G1 /S by double thymidine block and then released for 8 hours in nocodazole.41 GST (lane 1), GST-N (lane 2), GST-FAC (wild-type; lane 3), GST-FAC (L554P, lane 4), or GST-C1 (lane 5), prebound to glutathione-sepharose beads, was mixed with the extract. Bound cellular proteins were electrophoresed by SDS-PAGE and transferred to nitrocellulose. The filter was probed with either anti-GST monoclonal antibody or anti-cdc2 monoclonal antibody.

Close modal

Previous studies have suggested that FA cells have a defect in cell cycle progression, confined to the G2 /M transition phase.2,4,5 In the current manuscript, we show that expression of FAC protein is regulated during the cell cycle, with peak expression at the G2 /M transition point. Also, a 34-kD coprecipitating protein is observed, consistent with a previous report that FAC binds to a protein of 34 kD.50 We have shown that FAC binds to cdc2, which may well be one of the previously reported FAC-binding proteins. The binding interaction between FAC and cdc2 is mediated by the carboxyl terminus of FAC, a region important for functional activity of FAC in the cell. A FA patient-derived nonfunctional FAC polypeptide, with a L554P mutation in the carboxyl terminus, failed to bind to cdc2.

There are several cellular factors that may regulate the interaction between FAC and cdc2. Because FAC mRNA levels change minimally during cell cycle progression (data not shown), FAC protein levels appear to be regulated at a posttranscription level. Like other cdc2 binding proteins, such as cyclin B, FAC accumulates during S phase and is degraded during M phase. This similar pattern of degradation of cyclin B and FAC in mitosis may result from a common degradation mechanism, such as the anaphase promoting complex.51 Our data suggest that the FAC/cdc2 binding interaction is not regulated at the level of protein localization. FAC remains in the cytoplasm throughout the cell cycle. In addition, the binding interaction does not appear to be regulated by phosphorylation. There is no evidence for phosphorylation of FAC (Yamashita, unpublished observation, July 1994), and FAC does not contain consensus cdc2 phosphorylation sites. In contrast to FAC, unphosphorylated cdc2 levels remain constant during the cell cycle (Fig 1C), consistent with previous studies.32,46 Also, cdc2 is found in both the cytoplasm and the nucleus.52 Interestingly, the phosphorylation state and kinase activity of cdc2 are regulated during cell cycle progression. cdc2 is tyrosine phosphorylated on Y15 during S phase and dephosphorylated by cdc25 during the G2 /M transition.32-34,53,54 Taken together, our data suggest that the FAC/cdc2 interaction may be regulated by the amount of FAC in the cell or by the state of phosphorylation or activity of cdc2. Whether or not additional cellular proteins, such as cyclin B, bind to the FAC/cdc2 complex remains unknown.

The significance of the FAC/cdc2 binding interaction also remains unclear. Several potential cellular events resulting from the interaction are possible. In one model, FAC might be downstream of the cdc2 kinase. Once activated, cdc2 is known to phosphorylate several protein substrates, which mediate mitotic cellular events, including nuclear membrane disassembly and chromosome condensation.32 Consistent with this model, FAC may be a component of a mitotic DNA repair pathway downstream of active cdc2, similar to pathways observed in yeast.55-57 In an alternative model, FAC may bind and functionally regulate the activity of cdc2 by acting in concert with a cdk inhibitor that could couple a transient G2 /M pause with DNA repair. cdc2 exhibits persistent tyrosine phosphorylation after DNA damage, resulting in decreased cdc2/cyclin B kinase activity and cell cycle arrest.37,58,59 The interaction of cdc2 and FAC provides the opportunity for modulating this DNA damage-inducible checkpoint. In either case, a signal transduction cascade triggered by DNA damage might result, requiring interaction of cell cycle and DNA repair machinery at a G2 /M checkpoint.

The functional interaction between cdc2 and FAC also suggests that mutations in cdc2 could result in cancer susceptibility and an FA phenotype. The cdc2 gene (chromosome 10) does not map to known FA gene loci, but cdc2-like kinases and cdk inhibitors have been linked to tumor susceptibility.31,60,61 Accordingly, our data would suggest that the FAC pathway is inactive in cdc2 mutant cell lines.

Although the significance of the FAC/cdc2 protein interaction remains unclear, the interaction is consistent with the cell cycle abnormality of increased G2 percentage previously observed in FA cells.2,19,62 FAC levels and FAC/cdc2 interaction are maximal at the G2 /M transition. Accordingly, this is the point in the cell cycle at which FAC cells (lacking functional FAC protein) accumulate following crosslinking damage. Cell lines derived from FA patients with other types also accumulate abnormally at the G2 /M transition (Kupfer, unpublished observations, May 1996). As other FA genes are cloned, it will be interesting to investigate binding interactions between these proteins and the FAC/cdc2 complex.

We thank David Pellman, Bernard Mathey-Prevot, Chiang Li, and members of the D'Andrea laboratory for helpful discussions.

Supported in part by National Institutes of Health Grant No. R01-HL5725. G.M.K. is supported by Physician-Scientist Award No. K08-H103420, A.D.D. is a Scholar of the Leukemia Society of America (LSA), and D.N. is a Fellow of the LSA.

Address reprint requests to Gary M. Kupfer, MD, Division of Pediatric Oncology, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA 02115.

1
Fanconi
G
Familial constitutional panmyelocytopathy, Fanconi's anemia (FA). I. Clinical aspects.
Semin Hematol
4
1967
233
2
Strathdee CA, Buchwald M: Molecular and cellular biology of Fanconi anemia. Am J Pediatr Hematol/Oncol 14:177, 1992
3
Liu
J
Buchwald
M
Walsh
C
Young
N
Fanconi anemia and novel strategies for therapy.
Blood
84
1994
3995
4
Auerbach
AD
Fanconi anemia diagnosis and the diepoxybutane (DEB) test.
Exp Hematol
21
1993
731
5
Alter
BP
Fanconi's anaemia and its variability.
Br J Haematol
85
1993
9
6
Strathdee
CA
Duncan
AMV
Buchwald
M
Evidence for at least four Fanconi Anemia genes including FACC on chromosome 9.
Nat Genet
1
1992
196
7
Moustacchi
E
Papadopoulo
D
Diatloff-Zito
C
Buchwald
M
Two complementation groups of Fanconi's anemia differ in their phenotypic response to a DNA cross-linking treatment.
Hum Genet
75
1987
45
8
Duckworth-Rysiecki
G
Cornish
K
Clarke
CA
Buchwald
M
Identification of two complementation groups in Fanconi Anemia.
Somat Cell Mol Gen
11
1985
35
9
Joenje
H
LoTen
F
Oostra
A
Berkel
CV
Rooimans
M
Schroeder
S
Kurth
T
Wegner
R
Gille
J
Buchwald
M
Arwert
F
Classification of Fanconi Anemia patients by complementation analysis: Evidence for a fifth genetic subtype.
Blood
86
1995
2156
10
Strathdee
CA
Gavish
H
Shannon
W
Buchwald
M
Cloning of cDNAs for Fanconi's Anemia by functional complementation (Correction).
Nature
358
1992
434
11
Strathdee
CA
Gavish
H
Shannon
WR
Buchwald
M
Cloning of cDNAs for Fanconi's anaemia by functional complementation.
Nature
356
1992
763
12
The Fanconi Anaemia/Breast Cancer Consortium
Positional cloning of the Fanconi anaemia group A gene.
Nature Genetics
14
1996
324
13
Lo
Ten Foe JR
Rooimans
MA
Bosnoyan-Collins
L
Alon
N
Wijker
M
Parker
L
Lightfoot
J
Carreau
M
Callen
DF
Savoia
A
Cheng
NC
van Berkel
CGM
Strunk
MHP
Gille
JJP
Pals
G
Kruyt
FAE
Pronk
JC
Arwert
F
Buchwald
M
Joenje
H
Expression cloning of a cDNA for the major Fanconi anemia gene, FAA.
Nature Genetics
14
1996
320
14
Whitney
M
Thayer
M
Reifsteck
C
Olson
S
Smith
L
Jakobs
P
Leach
R
Naylor
S
Joenje
H
Grompe
M
Microcell mediated chromosome transfer maps the Fanconi anemia group D gene to chromosome 3p.
Nature Genet
11
1995
341
15
Chen
M
Tomkins
DJ
Auerbach
W
McKerlie
C
Youssoufian
H
Liu
L
Gan
O
Carreau
M
Auerbach
A
Groves
T
Guidos
CJ
Freeman
MH
Cross
J
Percy
DH
Dick
JE
Joyner
AL
Buchwald
M
Inactivation of Fac in mice produces inducible chromosomal instability and reduced fertility reminiscent of Fanconi anaemia.
Nature Genetics
12
1996
448
16
Whitney
MA
Royle
G
Low
MJ
Kelly
MA
Axthelm
MK
Reifsteck
C
Olson
S
Braun
RE
Heinrich
MC
Rathbun
RK
Bagby
GC
Grompe
M
Germ cell defects and hematopoietic hypersensitivity to gamma-interferon in mice with a targeted disruption of the Fanconi anemia C gene.
Blood
88
1996
49
17
Yamashita
T
Barber
DL
Zhu
Y
Wu
N
D'Andrea
AD
The Fanconi anemia polypeptide FACC is localized to the cytoplasm.
Proc Natl Acad Sci USA
91
1994
6712
18
Youssoufian
H
Localization of Fanconi anemia C protein to the cytoplasm of mammalian cells.
Proc Natl Acad Sci USA
91
1994
7975
19
Dutrillaux
B
Aurias
A
Dutrillaux
AM
Buriot
D
Prieur
M
The cell cycle of lymphocytes in Fanconi anemia.
Hum Genet
62
1982
327
20
Kaiser
TN
Lojewski
A
Dougherty
C
Juergens
L
Sahar
E
Latt
SA
Flow cytometric characterization of the response of Fanconi's Anemia cells to mitomycin C treatment.
Cytometry
2
1982
291
21
Digweed
M
Gunthert
U
Schneider
R
Seyschab
H
Friedl
R
Sperling
K
Irreversible repression of DNA synthesis in Fanconi anemia cells is alleviated by the product of a novel cyclin-related gene.
Mol Cell Biol
15
1995
305
22
Sabatier
L
Dutrillaux
B
Effect of caffeine in Fanconi anemia: I. Restoration of a normal duration of G2 phase.
Hum Genet
79
1988
242
23
Seyschab
H
Bretzel
G
Friedl
R
Schinlder
D
Sun
Y
Hoehn
H
Modulation of the spontaneous G2 phase blockage in Fanconi anemia cells by caffeine: Differences from cells arrested by X-irradiation.
Mutat Res
308
1994
149
24
Hain
J
Crompton
N
Burkart
W
Jaussi
R
Caffeine release of radiation induced S-phase and G2-phase arrest in V79 hamster cells-increase of histone messenger RNA levels and p34cdc2 activation.
Cancer Res
53
1993
1507
25
Diller
L
Kassel
J
Nelson
Ce
Gryka
MA
Wak
GL
Gebhardt
M
Bressac
B
Oxturk
M
Baker
SJ
Vogelstein
B
Friend
SH
p53 Functions as a cell cycle control protein in osteosarcomas.
Mol Cell Biol
10
1990
5772
26
Kastan
M
Zhan
Q
El-Deiry
W
Carrier
F
Jacks
T
Walsh
W
Plunkett
B
Vogelstein
B
AJ
Fornace J
A mammalian cell cycle checkpoint pathway utilizing p53 and GADD45 is defective in ataxiatalangiectasia.
Cell
71
1992
587
27
Kastan
M
Onyekwere
O
Sidransky
D
Vogelstein
B
Craig
R
Participation of p53 protein in the cellular response to DNA damage.
Cancer Res
51
1991
6304
28
Cross
SM
Sanchez
CA
Morgan
CA
Schimke
MK
Ramel
S
Idzerda
RL
Raskind
WH
Reid
BJ
A p53-dependent mouse spindle checkpoint.
Science
267
1993
1353
29
Kruyt
FAE
Dijkmans
LM
van den Berg
TK
Joenje
H
Fanconi Anemia genes act to suppress a cross-linker-inducible p53-independent apoptosis pathway in lymphoblastoid cell lines.
Blood
87
1996
938
30
Kupfer
GM
D'Andrea
AD
The effect of the Fanconi Anemia polypeptide, FAC, upon p53 induction and G2 checkpoint regulation.
Blood
88
1996
1019
31
Hirama
T
Koeffler
HP
Role of the cyclin-dependent kinase inhibitors in the development of cancer.
Blood
86
1995
841
32
Pines
J
Hunter
T
p34cdc2: The S and M kinase?
New Biologist
2
1990
389
33
Moreno
S
Hayles
J
Nurse
P
Regulation of p34cdc2 protein kinase during mitosis.
Cell
58
1989
361
34
Ducommun
B
Brambilla
P
Felix
MA Jr
Franza
BR Jr
Karsenti
E
Draetta
G
cdc2 phosphorylation is required for its interaction with cyclin.
EMBO J
10
1991
3311
35
Blangy
A
Lane
HA
d'Herin
P
Harper
M
Kress
M
Nigg
EA
Phosphorylation by p34cdc2 regulates spindle association of human Eg5, a kinesin-related motor essential for bipolar spindle formation in vivo.
Cell
83
1995
1159
36
Lock
RB
Ross
WE
Inhibition of p34cdc2 kinase activity by etoposide or irradiation as a mechanism of G2 arrest in Chinese hamster ovary cells.
Cancer Res
50
1990
3761
37
Nishio
K
Fujiwara
Y
Miyahara
Y
Takeda
Y
Ohira
T
Kubota
N
Ohta
S
Funayama
Y
Ogasawara
H
Ohata
M
Saijo
N
Cis-diamminedichloroplatinum(II) inhibits p34cdc2 protein kinase in human lung cancer cells.
Int J Cancer
55
1993
616
38
Herzinger
T
Funk
JO
Hillmer
K
Eick
D
Wolf
DA
Kind
P
Ultraviolet B irradiation-induced G2 cell cycle arrest in human keratinocytes by inhibitory phosphorylation of the cdc2 cell cycle kinase.
Oncogene
11
1995
2151
39
Tsao
Y-P
D'Arpa
P
Liu
LF
The involvement of active DNA synthesis in camptothecin-induced G2 arrest: Altered regulation of p34cdc2/Cyclin B.
Cancer Res
52
1992
1823
40
Lock
RB
Ross
WE
Possible role for p34cdc2 kinase in etoposide-induced cell death of Chinese hamster ovary cells.
Cancer Res
50
1990
3767
41
Rowley
R
Hudson
J
Young
PG
The wee1 protein kinase is required for radiation-induced mitotic delay.
Nature
356
1992
353
42
White
RJ
Gottlieb
TM
Downes
CS
Jackson
SP
Cell cycle regulation of RNA polymerase III.
Mol Cell Biol
15
1995
6653
43
Yamashita
T
Wu
N
Kupfer
G
Corless
C
Joenje
H
Grompe
M
D'Andrea
AD
The Clinical Variability of Fanconi Anemia (Type C) results from expression of an amino terminal truncated FAC polypeptide with partial activity.
Blood
87
1996
4424
44
D'Andrea
AD
Yoshimura
A
Youssoufian
H
Zon
LI
Koo
J-W
Lodish
HF
The cytoplasmic region of the erythropoietin receptor contains non-overlapping positive and negative growth regulatory domains.
Mol Cell Biol
11
1991
1980
45
Bradford
M
A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding.
Anal Biochem
72
1976
248
46
Draetta
G
Beach
D
Activation of cdc2 protein kinase during mitosis in human cells; cell cycle-dependent phosphorylation and subunit rearrangement.
Cell
54
1988
17
47
Gavish
H
dos Santos CC
Buchwald
M
A Leu554-to-Pro substitution completely abolishes the functional complementing activity of the Fanconi anemia (FACC) protein.
Hum Mol Genet
2
1993
123
48
Foe
LT
Rooimans
MA
Joenje
H
Arwert
F
A novel frameshift mutation (1806insA) in exon 14 of the Fanconi anemia C gene.
Hum Mutat
7
1996
264
49
Foe
LT
Barel
MT
Tuss
P
Digweed
M
Arwert
F
Joenje
H
Sequence variations in the Fanconi anaemia gene, FAC: Pathogenicity of 1806insA and R548X and recognition of D195V as a polymorphic variant.
Hum Genet
98
1996
522
50
Youssoufian
H
Auerbach
AD
Verlander
PC
Steimle
V
Mach
B
Identification of cytosolic proteins that bind to the Fanconi Anemia complementation group C polypeptide in vitro.
J Biol Chem
270
1995
9876
51
King
RW
Peters
JM
Tugendreich
S
Rolfe
M
Hieter
P
Kirschner
MW
A 20S complex containing CDC27 and CDC16 catalyzes the mitosis-specific conjugation of ubiquitin to cyclin B.
Cell
81
1995
279
52
Heald
R
McLoughlin
M
McKeon
F
Human wee1 maintains mitotic timing by protecting the nucleus from cytoplasmically activated cdc2 kinase.
Cell
74
1993
463
53
Hayles
J
Fisher
D
Woollard
A
Nurse
P
Temporal order of S phase and mitosis in fission yeast is determined by the state of the p34cdc2-mitotic B cyclin complex.
Cell
78
1994
813
54
Rime
H
Jessus
C
Ozon
R
Tyrosine phosphorylation of p34cdc2 is regulated by protein phosphatase 2A in growing immature Xenopus oocytes.
Exp Cell Res
219
1995
29
55
Montelone
BA
Koelliker
KJ
Interactions among mutations affecting spontaneous mutation, mitotic recombination, and DNA repair in yeast.
Curr Genet
27
1995
102
56
Kozhina
T
Kozhin
S
Stepanova
V
Yarovoy
B
Donich
V
Fedorova
I
Korolev
V
UVS112-a gene involved in excision repair of yeast.
Yeast
11
1995
1129
57
Heude
M
Chanet
R
Fabre
F
Regulation of Saccharomyces cerevisiae Srs2 helicase during the mitotic cell cycle, meiosis, and after irradiation.
Mol Gen Genet
248
1995
59
58
O'Connor
PM
Ferris
DK
Pagano
M
Draetta
G
Pines
J
Hunter
T
Longo
DL
Kohn
KW
G2 delay induced by nitrogen mustard in human cells affects cyclin A/cdk2 and Cyclin B1/cdc2-kinase complexes differently.
J Biol Chem
268
1993
8298
59
O'Connor
PM
Ferris
DK
White
GA
Pines
J
Hunter
T
Longo
DL
Kohn
KW
Relationships between cdc2 kinase, DNA cross-linking, and cell cycle perturbations induced by nitrogen mustard.
Cell Growth Differ
3
1992
43
60
MacLachlan
TK
Sang
N
Giordano
A
Cyclins, cyclin-dependent kinases and cdk inhibitors: Implications in cell cycle control and cancer.
Crit Rev Eukaryot Gene Expr
5
1995
127
61
Bullrich
F
MacLachlan
TK
Sang
N
Druck
T
Veronese
ML
Allen
SL
Chiorazzi
N
Koff
A
Heubner
K
Croce
CM
Giordano
A
Chromosomal mapping of members of the cdc2 family of protein kinases, cdk3, cdk6, PISSLRE, and PITALRE, and a cdk inhibitor, p27Kip1, to regions involved in human cancer.
Cancer Res
55
1995
1199
62
Kaiser
TN
Lojewski
A
Dougherty
C
Juergens
L
Sahar
E
Latt
SA
Flow cytometric characterization of the response of Fanconi's Anemia cells to mitomycin C treatment.
Cytometry
2
1982
291
Sign in via your Institution