Fanconi anemia (FA) is an autosomal recessive cancer susceptibility syndrome. The phenotype includes developmental defects, bone marrow failure, and cell cycle abnormalities. At least eight complementation groups (A-H) exist, and although three of the corresponding complementation group genes have been cloned, they lack recognizable motifs, and their functions are unknown. We have isolated a binding partner for the Fanconi anemia group C protein (FANCC) by yeast two-hybrid screening. We show that the novel gene, FAZF, encodes a 486 amino acid protein containing a conserved amino terminal BTB/POZ protein interaction domain and three C-terminal Krüppel-like zinc fingers. FAZF is homologous to the promyelocytic leukemia zinc finger (PLZF) protein, which has been shown to act as a transcriptional repressor by recruitment of nuclear corepressors (N-CoR, Sin3, and HDAC1 complex). Consistent with a role in FA, BTB/POZ-containing proteins have been implicated in oncogenesis, limb morphogenesis, hematopoiesis, and proliferation. We show that FAZF is a transcriptional repressor that is able to bind to the same DNA target sequences as PLZF. Our data suggest that the FAZF/FANCC interaction maps to a region of FANCC deleted in FA patients with a severe disease phenotype. We also show that FAZF and wild-type FANCC can colocalize in nuclear foci, whereas a patient-derived mutant FANCC that is compromised for nuclear localization cannot. These results suggest that the function of FANCC may be linked to a transcriptional repression pathway involved in chromatin remodeling.

FANCONI ANEMIA (FA) is an autosomal recessive disorder characterized by progressive pancytopenia, diverse congenital anomalies, and predisposition to cancer, particularly acute myeloid leukemia (AML) and squamous cell carcinoma.1-5 The diagnostic hallmark of FA cells is a unique hypersensitivity to DNA crosslinking agents such as mitomycin C (MMC) and diepoxybutane (DEB). In addition, FA cells have an abnormal cell cycle profile, described as an elongation or arrest at the G2 phase, and this abnormality is exacerbated by treatment with MMC.6,7 Because of associated genomic instability and cancer predisposition, FA has been classified along with xeroderma pigmentosum, Cockayne’s syndrome, trichothiodystrophy, ataxia telangiectasia, Bloom syndrome, Werner’s syndrome, and hereditary nonpolyposis colorectal cancer as one of the “caretaker gene diseases.”8 However, the basic defect in FA is unknown.

FA is genetically heterogeneous, comprising at least eight different complementation groups (A-H) identified by somatic cell hybrid analysis.9,10 Genes mutated in three of the FA complementation groups have been identified (FANCA,FANCC, and FANCG).11-13 In addition,FANCD has been mapped to chromosome 3p22-26,14 andFANCE has recently been mapped to chromosome 6p21-22.15 The proteins predicted to be encoded by the known FA genes lack recognizable motifs, and their functions are unknown. Because FA patients from different complementation groups have similar clinical and cellular phenotypes, a common molecular pathway in which the FA gene products participate has been hypothesized.

Although FA appears to have the characteristics of a DNA repair disorder, early work with the first known gene product, Fanconi anemia group C protein (FANCC), indicated that FANCC was located primarily in the cytoplasm, suggesting an indirect role in DNA repair.16,17 However, FANCC was subsequently also detected in the nuclear compartment, confirming that at least some FANCC is available in the nucleus for DNA-related activities.18,19Attempts to investigate even these most basic issues regarding subcellular localization of FANCC have been confounded by low endogenous expression levels, changes in expression during the cell cycle, and presence in both nuclear and cytoplasmic compartments.16-20 

It is likely that the activities of FA proteins are regulated or mediated by other cellular proteins. In support of this notion, evidence based on coimmunoprecipitation and in vitro binding experiments suggest that FANCC binds to several cytosolic proteins, including the molecular chaperone GRP94, nicotinamide adenine dinucleotide phosphate (NADPH) cytochrome P450 reductase, the cyclin dependent kinase cdc-2, and Fanconi anemia complementation group A (FANCA).18,20-22 Indeed, there is growing awareness that the FA proteins may interact with each other within a large protein complex.18,23,24 

We report here that FANCC interacts with a new human BTB/POZ (for Broad Complex, tramtrack, and Bric á Brac/pox virus and zinc finger) domain protein (for reviews see Albagli et al25 and Bardwell et al26), which we have named FAZF. The FAZF protein (for Fanconi anemia zincfinger) is similar to the promyelocytic zinc finger protein, PLZF, a Krüppel-like transcription factor involved chromosomal translocations with the retinoic acid receptor alpha (RARα) leading to acute promyelocytic leukemia (APL; for recent review, see Melnick et al27). In hematopoetic cells, PLZF is localized in nuclear speckles, which become delocalized in APL. PLZF represses transcription of specific targets by recruitment of histone deacetylase through the SMRT-mSin3-HDAC corepressor complex.28-32 In mammalian cells, FAZF is also located in nuclear speckles, which colocalize with wild-type FANCC, but not an inactivated FANCC protein. In addition, we show that FAZF is a transcriptional repressor and it readily forms heterodimers with PLZF. BTB/POZ proteins are implicated in oncogenesis,29,33-37hematopoiesis,38-40 and limb development,41-43suggesting that FANCC/FAZF interaction and the implied connection to transcriptional repression may be involved in the FA pathway.

Plasmids, cell lines, antibodies, and chemicals.

The bacterial expression plasmids for GST-FAN2 and GST-FAC1 have been described.19 To make the two-hybrid bait expressing FANCC amino acid #1-168 (pAS-FAN), polymerase chain reaction (PCR) primers (159-192 [5′-TTCGCTTTTTCCACCATGGCTCAAGATTCAGTAG-3′] and 660-704 [5′-GGGAGCCATTCGCCTTGGATCCTTCTATCCATTAAGATGATTCT-3′]) were used to amplify the FANCC sequence from a full-length cDNA contained in pLFACXN.19 The primers incorporatedNcoI and BamHI restriction sites in the amplified fragment for use in ligation. For p2HA-FANCC, full-length FANCC was obtained by PCR from pLFACXN with primers FAC5′ BamHI (5′-CGGGATCCGATGGCTCAAGATTCAGTAGATCTTTCT-3′) and FACW3′ BamHI (5′-CCGGATCCTAGACTTGAGTTCGCAGCTCTTTAAGGA-3′) and ligated into BamHI-digested/calf intestinal phosphatase (CIP)-treated pCEPHA2.RIG.G, a plasmid that encodes two tandem hemagglutinin (HA) epitopes (kindly provided by Dr Hans Joenje, Free University, Amsterdam, The Netherlands). 2HA-L554P was constructed similarly except that primer FACM3′ BamHI (5′-CCGGATCCTAGACTTGAGTTCGCGGCTCTTTAAGGA-3′) was used instead of FACW3 BamHI. For construction of the expression vector encoding the epitope-tagged pFlag-FAZF, total RNA was extracted from normal human peripheral blood, and primers FAZF5 BGL2ATC (5′-TACCCAAGCCAAGGCAAGATCTCAATGTCCCTGCCCCCCAT-3′) and FAZF3 BGL2 (5′-GCTACCGACACCCCGTAGATCTCAGGTGGTGGAGGAAGAA-3′) were used to obtain the cDNA by reverse transcription (RT)-PCR. The amplified fragment was digested withBglII, and ligated into BamHI-digested/CIP-treated vector pCEP4-Flag (kindly provided by Dr Naumovski, Stanford University, Stanford, CA). All PCR-derived sequences and ligation joints were verified by DNA dideoxy sequencing using an ABI PRISM Dye Terminator Cycle Sequencing Ready Reaction Kit (Perkin Elmer, Foster City, CA). Anti-HA was obtained from Roche Molecular Biochemicals (Indianapolis, IN). The monoclonal antibody (MoAb) specific for FANCC was produced by standard methods against the keyhole limpet hemacyanin-conjugated peptide C-ARELLKELRTQV, corresponding to FANCC aa #547-558.44 A detailed description of the antibody will be published elsewhere (M.E.H., in preparation). All chemicals were obtained from Sigma (St Louis, MO), unless otherwise indicated.

Two-hybrid library screen.

The two-hybrid screen was performed essentially as described previously.45 Briefly, Saccharomyces cerevisiae Y190 was transformed with the pAS-FAN bait (binding domain [BD] fusion) plasmid and selected for Trp prototrophy. After ensuring that the resulting strain expressed the FANCC bait and did not autoactivate the GAL4 reporter, it was transformed with a human B lymphocyte library encoding the activation domain (AD)–library hybrids. Approximately 300,000 His+ primary clones were obtained, and 59 of these were positive for β-galactosidase (gal) activity. Plasmid DNA prepared from several of the positive strains was transformed into Escherichia coli strain HB101, and transformants were selected. Candidate plasmids that did not repeat with the original bait, that autoactivated the reporter, or that interacted with unrelated baits were eliminated. The remaining interactors were further analyzed. A second screen was also performed with the same library using a BD expression vector encoding FANCC aa #116-558. For confirmation of two-hybrid interactions of PLZF and FAZF, the PJ69-4A strain of S cerevisiae46 was transformed with BD fusion constructs encoding full-length FAZF, PLZF, or the PLZF POZ domain. The transformed strains were selected on media lacking leucine, tryptophan, and adenine. Yeast colonies were counted and then selected in duplicate for liquid β-galactosidase assays as directed (Clontech, Palo Alto, CA). Results were normalized relative to the dimerization of PLZF. A full-length GAL4 plasmid was used as a positive control (Clontech). Other two-hybrid positive and negative control AD and BD domain plasmids were purchased from Clontech.

In vitro protein binding assays.

Expression and purification of Glutathione S-transferase (GST)-fusion proteins used in these assays have been described previously.19 Radiolabeled protein encoded by the library plasmid pAct8-1 was made by amplifying the insert using T7-promoter containing primers specific for flanking sequences in the pACT vector (Clontech). In vitro translations were performed using a commercial transcription-translation system (TnT, Promega, Madison, WI). Equal amounts of radiolabeled protein were added to GST fusion proteins immobilized on glutathione-Sepharose beads (GST, GST-FAC2, and GST-FAC1) and allowed to bind for 4 hours at 4°C in binding buffer (10 mmol/L Tris HCl/150 mmol/L NaCl/1 mmol/L EDTA/1% Nonidet P-40/1% deoxycholic acid). After extensive washing with the buffer, the beads were boiled in sample buffer and analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and autoradiography.

Northern blotting.

A multiple tissue blot of human poly (A+) RNA (Clontech) was probed with a radiolabeled 525-bp (BamHI/XhoI) restriction fragment from the library plasmid pAct8-1.

Immunoprecipitation and immunoblotting.

Cells were washed twice with phosphate-buffered saline (PBS), and cell lysates were solubilized in lysis buffer 1 (20 mmol/L Tris-Cl [pH 7.5], 150 mmol/L NaCl, 1% Triton X-100, 10 mmol/L EDTA, 1% deoxycholate, 1.5% aprotinin, 1 mmol/L phenylmethylsulfonyl fluoride [PMSF]). Anti-Flag M2-affinity gel (Sigma) or anti-HA (Roche Molecular Biochemicals) followed by Protein A/G PLUS-Agarose beads (Santa Cruz Biotechnology, Santa Cruz, CA) was added as indicated in the text, and the sample was rocked overnight at 4°C. The immune complexes were sedimented by centrifugation, washed in lysis buffer, mixed with Laemmli sample buffer, boiled, and separated by SDS-PAGE. Proteins were electroblotted onto Bio-Blot nitrocellulose (Costar, Cambridge, MA) and probed with anti-HA, anti-Flag M2 (Sigma), or anti-FANCC MoAb 3A11 as previously described.19 For the PLZF-FAZF interaction, cells were lysed in lysis buffer 2 (1% NP40, 50 mmol/L NaCl, 50 mmol/L Tris pH 8.0, 1 mmol/L MgCl2, 10 mmol/L ZnCl2, 4% glycerol and Complet protease inhibitors [Roche Molecular Biochemicals]). Immune complexes were obtained essentially as described above using anti-Flag MoAb (Kodak, Rochester, NY) and Protein A agarose (Roche Molecular Biochemicals). The electrophoresed proteins were transferred to Immobilon PVDF membrane (Millipore, Bedford, MA) and were processed as described above using anti-PLZF polyclonal antibody.47 

Transient transfection and immunofluorescence microscopy.

293 Epstein-Barr nuclear antigen (EBNA) cells (Invitrogen, Carlsbad, CA) were grown on chamber slides and transfected using lipofectamine as directed by the manufacturer (GIBCO, Grand Island, NY). Plasmids encoding epitope-tagged FAZF, FANCC, L554P, or parental vectors as negative controls for antibody specificity were used as indicated in text. Cells were processed as described previously19 using monoclonal anti-HA 12CA5 (Roche Molecular Biochemicals) and/or rabbit polyclonal anti-Flag (Zymed, South San Francisco, CA) and Oregon Green–conjugated goat antimouse and/or Texas Red–conjugated goat antirabbit (Molecular Probes, Eugene, OR) as secondary antibodies. Double-labeled images were acquired with a Bio-Rad MRC 1024 ES laser scanning confocal imaging system (Bio-Rad Laboratories, Richmond, CA) attached to an inverted Nikon Eclipse TE300 microscope (Nikon, Melville, NY). The acquisition system (LaserSharp, Hercules, CA) uses a krypton/argon laser with excitation lines at 488 and 564 nm, and sequential detection using three 8-bit photomultiplier tubes (PMTs). Single-labeled images were acquired with a Leica 900 confocal laser-scanning microscope (Leica Inc, Deerfield, IL) equipped with a krypton/argon detector. Collected images were imported into Adobe Photoshop 4.0 (Adobe, San Jose, CA) and overlapped to produce merged images. Localization study data (see Table 2) was gathered with a Leitz Orthoplan 2 fluorescence microscope (Ernst Leitz GMBH, Wetzlar, Germany) using green (488 nm) or red (568 nm) filters and a ×100 oil lens. One hundred cells expressing the protein of interest were counted for each cell line. Cells were scored as positive if they contained two or more nuclear foci. Standard error was computed using StatView 5.0 (SAS Institute, Cary, NC).48 

Electrophorectic mobility shift assay (EMSA).

293T (ATCC No. CRL1573) cells were transfected with 20 μg of DNA using the Ca PO4 method.49 Nuclear extracts were prepared 48 hours after transfection, frozen in liquid nitrogen, and stored at −70°C.50 Synthetic duplexes (Table 1) were end-labeled using the large Klenow fragment of E coli DNA polymerase and [α-P32] dCTP (3,000 Ci/mmol), and purified by spin column or PAGE. Each binding reaction contained approximately 2 mg nuclear extract protein in 20 mmol/L HEPES pH 7.5, 1 mmol/L MgCl2, 10 mmol/L ZnCl2, 4% glycerol, 100 mg/mL bovine serum albumin (BSA), and 1 mg dIdC, and was incubated on ice for 30 minutes. Unlabeled oligonucleotide competitors or anti-Flag antibody (Kodak) was added at least 20 minutes before the addition of 10 femtomoles of labeled duplex as indicated in the text. After a further incubation of 20 minutes, protein-DNA complexes were separated by electrophoresis on a 4% nondenaturing polyacrylamide (30:1, acrylamide:bis-acrylamide) gel.

Table 1.

Transcriptional Target Sites

Site A  
 5′-TCGAGCTCTCGCTTTGTCAAGATACAGTTCAAAGCACTG-3′ 
  3′-GAGAGCGAAACAGTTCTATGTCAAGTTTCGTGACGAGCT-5′  
Site B 
 5′-TCGAGGTATTCAGTACAGTACCAT-3′ 
  3′-CATAAGTCATGTCATGGTAGAGCT-5′  
Lex 
 5′-TCGAGAGTGGTTATATGTACAGTACTCGAGG-3′ 
  3′-TCACCAATATACATGTCATGAGCTCCGAGCT-5′  
IL-3R 
 5′-TCGAGGATGACTGCGAGTACAGTTGCAACGG-3′ 
  3′-CTACTGACGCTCATGTCAACGTTGCCGAGCT-5′  
IL-3Rmut 
 5′-TCGAGGATGACTGCGAGTAAAGTTGCAACGG-3′ 
  3′-CTACTGACGCTCATTTCAACGTTGCCGAGCT-5′ 
Site A  
 5′-TCGAGCTCTCGCTTTGTCAAGATACAGTTCAAAGCACTG-3′ 
  3′-GAGAGCGAAACAGTTCTATGTCAAGTTTCGTGACGAGCT-5′  
Site B 
 5′-TCGAGGTATTCAGTACAGTACCAT-3′ 
  3′-CATAAGTCATGTCATGGTAGAGCT-5′  
Lex 
 5′-TCGAGAGTGGTTATATGTACAGTACTCGAGG-3′ 
  3′-TCACCAATATACATGTCATGAGCTCCGAGCT-5′  
IL-3R 
 5′-TCGAGGATGACTGCGAGTACAGTTGCAACGG-3′ 
  3′-CTACTGACGCTCATGTCAACGTTGCCGAGCT-5′  
IL-3Rmut 
 5′-TCGAGGATGACTGCGAGTAAAGTTGCAACGG-3′ 
  3′-CTACTGACGCTCATTTCAACGTTGCCGAGCT-5′ 
Transcriptional repression assays.

Plasmids encoding PLZF in the pSG5 expression vector were previously described.51 The Flag-epitope tagged version of FAZF was constructed in the pSG5 vector by standard methods. Four copies of a PLZF binding site found within the interleukin (IL)-3 receptor promoter (5′-TCGAGGATGACTGCGAGTACAGTTGCAACGG-3′)51a were cloned 5′ of the thymidine kinase promoter driving transcription of a luciferase reporter gene (IL-3R-Luc). For transcription assays, 2 × 105 293T cells per well of a 12-well dish were transfected with 1 μg of DNA, or as indicated in text, and 5 μL of Superfect (Qiagen, Valencia, CA). Luciferase levels were measured 48 hours after transfection using a Dual Luciferase kit (Promega).

Identification of a FANCC-interacting protein by yeast two-hybrid screening.

FANCC has been shown to bind to other cellular proteins and may be part of a large intracellular complex.18,20-22,24,52 We used a yeast two-hybrid assay to screen for proteins that bind to FANCC in vivo in an effort to identify binding partners that might shed light on the function of the FA pathway.45 The bait was designed (Fig 1A) to contain a region of FANCC deleted in patients with a severe phenotype (IVS4-4A-T), reasoning that this may be an area for functionally important protein-protein contact.14,53 Related strategies to compare strains expressing wild-type and mutant proteins (eg, full-length FANCC baits, and the inactive L554P protein) were unsuccessful because the strains were unstable and could not be used for screening (data not shown). The bait strain containing N-terminal sequences (FAN, aa #1-168) was used to screen a human B-cell library.45 Transformants were selected that were HIS+, and β-gal+, that did not autoactivate the reporter, interacted with the FAN bait on retesting, and did not have reporter activity with unrelated baits. Figure 1B shows the two-hybrid interaction of a representative interacting clone encoded by library plasmid pACT8-1. Southern blot analysis of candidates from the screen using an internal fragment from clone 8-1 indicated that at least three clones of different sizes contained hybridizing sequences, suggesting that the same candidate had been selected in the screen several times. Additional screening performed with a FANCC bait containing aa #116-558 also resulted in capturing sequences contained in clone 8-1, although this bait proved to be too unstable for further analysis (data not shown). DNA sequence analysis of the hybridizing clones indicated that the clones contained overlapping regions of the same sequence (data not shown).

Fig. 1.

Identification of a FANCC-interacting protein by two-hybrid screening. (A) FANCC two-hybrid bait. The upper open rectangle represents wild-type FANCC. Asterisks indicate location of the R548X and L554P inactivating mutations.11,53,82 The lower open rectangle represents the region of FANCC expressed as a two-hybrid bait. The numbers indicate the corresponding amino acid residue of FANCC (the binding domain of the two-hybrid fusion is not shown for simplicity). The overlap with wild-type is indicated by the arrangement of the boxes in which the shaded area corresponds to the deletion in the IVS4 4A-T inactivating mutation that leads to a severe disease phenotype.14,53 (B) (see page 3740) Two-hybrid interaction. The Y190 yeast strain was cotransformed with the FAN bait plasmid and a representative prey plasmid (8-1) isolated by two-hybrid screening. Controls are shown for strains containing the prey plasmid 8-1 and an unrelated bait, and strains known to be positive and negative for interaction. The cotransformed strains are shown growing on media selective for both bait and prey plasmids (-Trp, -Leu), and under HIS selection. The lower panel shows results of the β-gal assay. (C) The protein encoded by clone 8-1 interacts with an amino terminal region of FANCC in vitro. Radiolabeled protein made by coupled in vitro transcription and translation from 8-1 cDNA bound specifically to immobilized GST-FANC1, but not to immobilized GST or GST-FAN2.

Fig. 1.

Identification of a FANCC-interacting protein by two-hybrid screening. (A) FANCC two-hybrid bait. The upper open rectangle represents wild-type FANCC. Asterisks indicate location of the R548X and L554P inactivating mutations.11,53,82 The lower open rectangle represents the region of FANCC expressed as a two-hybrid bait. The numbers indicate the corresponding amino acid residue of FANCC (the binding domain of the two-hybrid fusion is not shown for simplicity). The overlap with wild-type is indicated by the arrangement of the boxes in which the shaded area corresponds to the deletion in the IVS4 4A-T inactivating mutation that leads to a severe disease phenotype.14,53 (B) (see page 3740) Two-hybrid interaction. The Y190 yeast strain was cotransformed with the FAN bait plasmid and a representative prey plasmid (8-1) isolated by two-hybrid screening. Controls are shown for strains containing the prey plasmid 8-1 and an unrelated bait, and strains known to be positive and negative for interaction. The cotransformed strains are shown growing on media selective for both bait and prey plasmids (-Trp, -Leu), and under HIS selection. The lower panel shows results of the β-gal assay. (C) The protein encoded by clone 8-1 interacts with an amino terminal region of FANCC in vitro. Radiolabeled protein made by coupled in vitro transcription and translation from 8-1 cDNA bound specifically to immobilized GST-FANC1, but not to immobilized GST or GST-FAN2.

Close modal

To confirm the in vivo interactions we found in yeast, we tested a radiolabeled in vitro translated protein encoded by clone 8-1 for binding to bacterially expressed GST-FANCC fusion proteins immobilized on glutathione Sepharose. In these protein affinity experiments, radiolabeled 8-1 protein bound to immobilized GST-FAC1 (aa #106-558), but not to GST-FAN2 (aa #7-106) or to GST alone (Fig 1C), indicating that the binding between the 8-1 protein and FANCC is direct and specific for the amino terminal region of FANCC. Because the FAN yeast bait contains aa #1-168, the in vitro and in vivo binding data suggest that the region of FANCC participating in the interaction is approximately between aa #106-168. Thus, binding between protein encoded by clone 8-1 and FANCC corresponds roughly to the deletion beginning at aa #116, resulting from the IVS4-4A-T mutation.54 

Characterization of FANCC interacting clone 8-1.

PCR primers were used to amplify the inserts from the library plasmid pAct8-1. DNA sequence analysis revealed an open reading frame of 300 amino acids in-frame with two-hybrid activation domain sequences. A BLASTX database search (http://www.ncbi.nlm.nih.gov/BLAST/)55,56 with the 8-1 sequence indicated similarities to multiple zinc finger–containing proteins, and near identity with sequences contained within exons of a genomic clone from human chromosome 19q13.1 (cosmid F24109, GenBank accession No. AD000671). Thus, FAZF does not map to the location of a mapped FA gene. This cosmid clone contained additional 5′ sequences, suggesting that clone 8-1 was not complete. Therefore, the 1,461-bp full-length FAZF was cloned by RT-PCR using total RNA from normal human peripheral blood. Recently, another group has also deposited sequences in agreement with our data (accession no. AF130255, unpublished).

Examination of the predicted FAZF protein sequence indicated a significant degree of homology with the PLZF protein of t(11;17) (q21;q23)–associated APL (reviewed in Melnick et al27; Fig 2A). FAZF contains an amino terminal POZ/BTB domain25,57 sharing highly conserved residues appearing in this subfamily that includes LRF,41LAZ-3/BCL-6,33,36,37 and the recently cloned Kaiso.58 The POZ domain can mediate protein dimerization59,60 and transcriptional repression by PLZF.61 PLZF binds to specific DNA sequences and can do so using only the last five to seven of nine zinc finger motifs.61,62 FAZF’s three zinc fingers are closest in homology (91% similar) to the last in the series of nine at the C-terminus of PLZF (Fig 2B), and the POZ/BTB domain is nearly 60% similar. The degree of similarity between PLZF and FAZF suggested that these proteins might have common transcriptional functions. Examination of the genomic database also indicated a possible evolutionary relationship between PLZF and FAZF. A search of the EST database (expressed sequence tags; NCBI/NLM, Bethesda, MD) showed that the FAZF sequence (UniGene Hs. 99430;http://www.ncbi.nlm.nih.gov/UniGene/) is contained in a clone that also includes the gene HRX2, a gene very similar to theMLL/HRX (mixed lymphoid or mixed lineage leukemia/homologue of trithorax) gene,63-66 located adjacent to the PLZF gene on chromosome 11. This finding suggests that the simpler FAZF gene with its three zinc fingers might be an evolutionary antecedent to PLZF. The central portion of FAZF has no significant homology to PLZF or any other sequences in the database. The lack of homology in sequences between the BTB/POZ domain and the zinc fingers is a common feature in other BTB/POZ and Krüppel-like proteins. The FAZF sequences between aa #1-98, including part of the BTB/POZ domain (aa #1-109), are apparently not required for FANCC interaction, because we obtained only clones missing the amino terminal portion of the protein in the two-hybrid screens.

Fig. 2.

FAZF is similar to PLZF. (A) Comparison of the amino acid sequences of FAZF and PLZF showing BTB/POZ domains (underlined at the amino termini) and zinc fingers (underlined at the C-termini) in CLUSTAL W format. The FAZF sequence has been submitted to GenBank (accession no. AF165097). (B) Schematic representation of the PLZF and FAZF proteins indicating regions of homology and similarity.

Fig. 2.

FAZF is similar to PLZF. (A) Comparison of the amino acid sequences of FAZF and PLZF showing BTB/POZ domains (underlined at the amino termini) and zinc fingers (underlined at the C-termini) in CLUSTAL W format. The FAZF sequence has been submitted to GenBank (accession no. AF165097). (B) Schematic representation of the PLZF and FAZF proteins indicating regions of homology and similarity.

Close modal

A Northern blot survey of RNA from multiple tissues probed with a radiolabeled restriction fragment from clone 8-1 revealed a predominant weak transcript of 1.4 kb in most tissues, except for testis, in which multiple strong transcripts were observed (2.0, 2.7, and 4.4 kb) and the 1.4-kb transcript was absent. Transcripts present in testis, particularly the 4.4-kb band, were also present in other tissues as minor transcripts. A strong signal was observed in peripheral blood leukocytes, consistent with the presence of FAZF hybridizing sequences in the mRNA from cells similar to that used for construction of the two-hybrid expression library (Fig 3).

Fig. 3.

FAZF is ubiquitously expressed. Multiple human tissue northern blot (Clontech) was probed using radiolabeled restriction fragment from clone 8-1. Lane 1, spleen; lane 2, thymus; lane 3, prostate; lane 4, testis; lane 5, ovary; lane 6, small intestine; lane 7, colon; lane 8, peripheral blood mononuclear leukocyte. Longer exposure of this blot showed a 1.4-kb FAZF band in all tissues except testis.

Fig. 3.

FAZF is ubiquitously expressed. Multiple human tissue northern blot (Clontech) was probed using radiolabeled restriction fragment from clone 8-1. Lane 1, spleen; lane 2, thymus; lane 3, prostate; lane 4, testis; lane 5, ovary; lane 6, small intestine; lane 7, colon; lane 8, peripheral blood mononuclear leukocyte. Longer exposure of this blot showed a 1.4-kb FAZF band in all tissues except testis.

Close modal
FAZF coimmunoprecipitates with FANCC.

To determine whether FANCC associates with FAZF in vivo, expression vectors for epitope-tagged FANCC and full-length FAZF were cotransfected into 293 EBNA cells. Immobilized anti-Flag MoAb was added to lysates prepared from an asynchronous population of transfected cells, and the resulting complexes were separated by SDS-PAGE and analyzed by immunoblotting with anti-Flag. Whole cell extracts (WCE) were analyzed as controls for protein expression. As shown in Fig 4B, Flag-FAZF is detected as an Mr = 56,000 protein in whole cell extracts (lanes 2 and 5) and in lysates first immunoprecipitated with anti-Flag (lanes 3 and 4). The Flag-FAZF signal is absent in lysates from cells transfected with the parental vector (lane 7), or in lysates from cells expressing HA-FANCC (lane 1). We conclude that FAZF is expressed, is approximately the size expected, and is specifically detected by immunoprecipitation and by simple blotting in the whole cell lysates of cells transfected with the Flag-FAZF expression vector.

Fig. 4.

FANCC and full-length FAZF interact in vivo. 293 EBNA cells were transiently transfected with epitope-tagged expression vectors for HA-FANCC and/or Flag-FAZF. Lysates were electrophoresed directly as WCE or after immunoprecipitation with anti-Flag epitope tag. Proteins were detected by immunoblotting with anti-Flag or anti-FANCC. (A) Lane 1, FANCC WCE; lane 2, FANCC + FAZF WCE; lane 3, FANCC + FAZF IP; lane 4, FAZF IP; lane 5, FAZF WCE; lane 6, FANCC IP; lane 7, pCEP-Flag WCE. (B) The same loading order as in (A), probed with anti-Flag.

Fig. 4.

FANCC and full-length FAZF interact in vivo. 293 EBNA cells were transiently transfected with epitope-tagged expression vectors for HA-FANCC and/or Flag-FAZF. Lysates were electrophoresed directly as WCE or after immunoprecipitation with anti-Flag epitope tag. Proteins were detected by immunoblotting with anti-Flag or anti-FANCC. (A) Lane 1, FANCC WCE; lane 2, FANCC + FAZF WCE; lane 3, FANCC + FAZF IP; lane 4, FAZF IP; lane 5, FAZF WCE; lane 6, FANCC IP; lane 7, pCEP-Flag WCE. (B) The same loading order as in (A), probed with anti-Flag.

Close modal

To determine if FANCC was coimmunoprecipitated with Flag-FAZF, the blot was stripped and reprobed with an antibody specific for FANCC (Fig 4A). In cell lysates containing Flag-FAZF, FANCC was detected (lane 3), and this band comigrated with the HA-FANCC in lanes 1 and 2 (whole cell lysates containing HA-FANCC and both HA-FANCC and Flag-FAZF, respectively). We conclude that HA-FANCC coimmunoprecipitates with Flag-FAZF. The reverse experiment was performed on the same lysates by first immunoprecipitating HA-FANCC with anti-HA serum followed by immunoblotting with either anti-FANCC or anti-Flag. Although the background was much higher using the HA antibody, we found that Flag-FAZF was specifically coimmunoprecipitated with HA-FANCC (data not shown).

FAZF is a nuclear protein with a punctate pattern.

To determine the subcellular localization of FAZF, a Flag epitope-tagged protein was expressed in 293 EBNA cells and analyzed by indirect immunofluorescence. Cells were transfected with pFlag-FAZF or pFlag parental vector (as a negative control for specificity of the Flag antisera) and processed 20 hours later for confocal microscopy. Figure 5 (see page 3740) shows representative views of FAZF expression patterns. Consistent with observations reported for PLZF and other BTB/POZ-containing proteins, FAZF is almost entirely nuclear, with most cells containing foci superimposed on a microspeckled nuclear pattern.26,40,47,60,67 Like PLZF foci, FAZF nuclear foci varied in size and number, but were present in nearly all cells observed (Table 2). Staining was absent in cells transfected with pFlag.

Fig. 5.

FANCC and FAZF colocalize in nuclear bodies. 293 EBNA cells were transfected with pFlag-FAZF, pHA-FANCC, or pHA-L554P, or the pFlag parental vector (control panel), as indicated. The fixed and permeabilized cells were stained with antibodies specific for the epitope tags, followed by staining with fluorescent-conjugated secondary antibodies (Red = FAZF and Green = FANCC or L554P). Immunofluoresence was examined by confocal laser microscopy. For double labeling, the green and red channels were recorded independently and then overlaid. (A) Subcellular localization of FAZF. Original magnification ×63. (B) Colocalization of FANCC and FAZF in nuclear foci. Original magnification ×60. (C) The mutant L554P protein does not colocalize with FAZF.

Fig. 5.

FANCC and FAZF colocalize in nuclear bodies. 293 EBNA cells were transfected with pFlag-FAZF, pHA-FANCC, or pHA-L554P, or the pFlag parental vector (control panel), as indicated. The fixed and permeabilized cells were stained with antibodies specific for the epitope tags, followed by staining with fluorescent-conjugated secondary antibodies (Red = FAZF and Green = FANCC or L554P). Immunofluoresence was examined by confocal laser microscopy. For double labeling, the green and red channels were recorded independently and then overlaid. (A) Subcellular localization of FAZF. Original magnification ×63. (B) Colocalization of FANCC and FAZF in nuclear foci. Original magnification ×60. (C) The mutant L554P protein does not colocalize with FAZF.

Close modal
Table 2.

Formation of Nuclear Foci for FAZF, FANCC, and L554P

Protein % of Expressing Cells With 2+ Nuclear Foci
FAZF  93% ± 2.6%  
FANCC  24% ± 4.3%  
FANCC (L554P)  0%* ± 0% 
Protein % of Expressing Cells With 2+ Nuclear Foci
FAZF  93% ± 2.6%  
FANCC  24% ± 4.3%  
FANCC (L554P)  0%* ± 0% 
*

One foci was found in each of 2 cells of 100 counted.

Wild-type but not mutant FANCC colocalizes with FAZF in nuclear bodies.

To determine if FAZF and FANCC proteins colocalize, we performed experiments using indirect immunofluorescence and confocal microscopy to examine cells that were doubly transfected with constructs encoding tagged FAZF and FANCC. A survey of cells expressing FANCC (either singly or coexpressed with FAZF) showed that FANCC is distributed in both cytoplasmic and nuclear compartments, as reported previously.16-19 In addition, we observed that the distribution of FANCC between cytoplasm and nucleus varied, and that among cells expressing FANCC in an asynchronous population, 24% of cells contained two or more nuclear foci (Table 2 and Fig 5B, FANCC panel). The amount of FANCC appearing in the nucleus, or in foci, did not seem to be correlated with higher expression levels. Indeed, in many cells where FANCC expression was highest, no FANCC was observed in the nuclear compartment at all (data not shown). In cells that contained foci, the typical number ranged from approximately 5 to 20. The foci were typically superimposed on a diffuse nuclear stain, which was excluded from the nucleoli. Table 2 summarizes the percentage of cells containing FANCC foci observed in this experiment. Similar results were obtained from a 293 cell line stably expressing FANCC without an epitope tag using a FANCC-specific MoAb (data not shown). Interestingly, a similarly complex and variable pattern was recently described for another FA protein, FANCA, by D’Andrea et al,68 suggesting that foci formation may be an important and newly appreciated feature of FANCC and FANCA subnuclear localization. Not all cells expressing FANCC contain foci, as described above, but when FAZF and FANCC foci did appear in the same cell, we observed extensive overlap. Figure 5C shows a representative cell in which there is overlap of nuclear foci. No colocalization was observed between FANCC and PLZF, or between promyelocytic leukemia gene product (PML) and FANCC in colocalization experiments performed similarly (data not shown).

Although the FAZF interaction site on FANCC appears to be between aa #106 and 168, well away from the C-terminus of the FANCC, we wondered whether the L554P mutant protein could colocalize with the FAZF nuclear foci, because other features of the defective protein besides this interaction might influence colocalization. To answer this question, we expressed an HA-epitope–tagged L554P protein in 293 EBNA cells and observed the staining pattern of L554P with and without FAZF coexpression. In contrast with the pattern observed for wild-type FANCC, L554P staining shows intense, diffuse cytoplasmic staining with minimal nuclear staining (Fig 5C, L554P panel). In addition, although the expression of L554P was comparable with that of wild-type FANCC, nuclear foci were very rare; of 100 cells expressing L554P, only one foci in each of two cells was observed (Table 2), suggesting that the ability of L554P to enter the nucleus is compromised under these conditions. A recent report by Savoia and others described similar observations for L554P overexpressed in Hela cells.69 When L554P and FAZF are expressed in the same cells, the proteins are localized to different parts of the cells (Fig 5C [overlap]). In the cell shown, the FAZF staining shows a micropunctate nuclear staining pattern, and the L554P staining is mainly cytoplasmic. When these two photos were overlaid, no colocalization was observed. No FAZF/L554P colocalization was observed in other cells examined, although other cells coexpressing L554P and FAZF contained larger FAZF foci (data not shown) consistent with FAZF’s overall variable nuclear staining pattern.

FAZF binds to PLZF target sequences.

The striking homology between PLZF and FAZF suggested that the two proteins may have similar functions. PLZF has been shown to bind to specific DNA target sequences.61 To determine if FAZF could bind to the same sequences as PLZF, we performed an EMSA with nuclear extracts of 293T cell transfected with the PLZF or FAZF expression constructs, using a radiolabeled high-affinity PLZF binding site (Table1, Site B) derived from a CpG island library.51aFigure 6A shows that both PLZF and FAZF formed DNA-protein complexes with this site, indicated by the appearance of shifted bands in lanes containing FAZF or PLZF extracts. No shifted bands appear in the control lanes where no extracts, or mock transfected extracts, were added. The complexes caused by the epitope-tagged FAZF protein could be supershifted by the addition of an anti-Flag antibody (Fig 6A, right panel), confirming that the shifted band contains tagged FAZF. In a competition assay, the PLZF- and FAZF-DNA complexes were abrogated by addition of a molar excess of unlabeled site B, a high affinity site from the IL-3 receptor promoter (Fig 6B) as well as the Lex operator (data not shown), which is also a target of PLZF.62 A missense mutant within the IL3R site abrogated binding to both PLZF and FAZF. Together, these data suggest that PLZF and FAZF may bind the same or a closely overlapping set of target genes.

Fig. 6.

FAZF and PLZF bind to the same DNA sequences. (A) Nuclear extracts from untransfected cells (control) or from 293T cells transfected with a PLZF expression vector or a Flag-FAZF expression vector were allowed to interact with a high affinity PLZF binding site (Site B, Table 1), followed by nondenaturing gel electrophoresis. A supershift assay of the Flag-FAZF complexes was performed by the addition of Flag M2 MoAb to the reaction. (B) Competition analysis of FAZF and PLZF DNA binding. Extracts containing PLZF or FAZF were allowed to bind to a high affinity PLZF site (Site B, Table 1) in the absence of competitor or in the presence of a 10-fold, 100-fold, or 1,000-fold molar excess of the indicated unlabeled competitor oligonucleotide. The DNA protein complexes were resolved as described above.

Fig. 6.

FAZF and PLZF bind to the same DNA sequences. (A) Nuclear extracts from untransfected cells (control) or from 293T cells transfected with a PLZF expression vector or a Flag-FAZF expression vector were allowed to interact with a high affinity PLZF binding site (Site B, Table 1), followed by nondenaturing gel electrophoresis. A supershift assay of the Flag-FAZF complexes was performed by the addition of Flag M2 MoAb to the reaction. (B) Competition analysis of FAZF and PLZF DNA binding. Extracts containing PLZF or FAZF were allowed to bind to a high affinity PLZF site (Site B, Table 1) in the absence of competitor or in the presence of a 10-fold, 100-fold, or 1,000-fold molar excess of the indicated unlabeled competitor oligonucleotide. The DNA protein complexes were resolved as described above.

Close modal
FAZF interacts with PLZF.

Because BTB/POZ domain-containing proteins have been known to form functional dimers,26,59 we next determined whether PLZF and FAZF might physically interact. PLZF and FAZF combinations were coexpressed as AD/BD fusion proteins in yeast, and assayed for reporter activity. We found that PLZF protein self-associated in this assay, as expected, and the POZ domain of PLZF (POZ-PLZF) was sufficient for this interaction (Fig 7A). FAZF (AD) could similarly interact with a BD (full-length) PLZF or BD-POZ-PLZF. We conclude that the PLZF POZ domain is sufficient for interaction of PLZF and FAZF. To confirm the interaction observed in yeast, we tested for PLZF-FAZF interaction by coimmunoprecipitation in mammalian cells. In 293T cells cotransfected with expression vectors encoding PLZF and a Flag-tagged FAZF expression vector, PLZF could be detected after immunoprecipitation of the epitope-tagged FAZF (Fig 7B).

Fig. 7.

PLZF and FAZF interact in vivo. (A) Yeast two-hybrid assay. PJ69-4A yeast were transformed with a bait plasmid encoding full-length PLZF or the POZ domain of PLZF linked to the GAL4 DNA binding domain and the indicated prey plasmids containing full-length FAZF or PLZF or the POZ domain of PLZF linked to an acidic activation domain. Yeast colonies grown on leucine and tryptophan deficient media were expanded, and a liquid β-galactosidase assay was performed. Levels of β-galactosidase were normalized, setting the level of β-galactosidase obtained in the presence of a PLZF bait and PLZF prey plasmid to 1.0. The GAL4 protein acted as positive control for transcription. (B) In vivo–coimmunoprecipitation. 293T cells were transfected with PLZF or Flag-FAZF plasmids as indicated. Lysates from the cells were subjected to immunoprecipitation with the Flag M2 MoAb followed by immunoblotting with a PLZF polyclonal antibody.47 

Fig. 7.

PLZF and FAZF interact in vivo. (A) Yeast two-hybrid assay. PJ69-4A yeast were transformed with a bait plasmid encoding full-length PLZF or the POZ domain of PLZF linked to the GAL4 DNA binding domain and the indicated prey plasmids containing full-length FAZF or PLZF or the POZ domain of PLZF linked to an acidic activation domain. Yeast colonies grown on leucine and tryptophan deficient media were expanded, and a liquid β-galactosidase assay was performed. Levels of β-galactosidase were normalized, setting the level of β-galactosidase obtained in the presence of a PLZF bait and PLZF prey plasmid to 1.0. The GAL4 protein acted as positive control for transcription. (B) In vivo–coimmunoprecipitation. 293T cells were transfected with PLZF or Flag-FAZF plasmids as indicated. Lysates from the cells were subjected to immunoprecipitation with the Flag M2 MoAb followed by immunoblotting with a PLZF polyclonal antibody.47 

Close modal
FAZF is a transcriptional repressor.

To determine if FAZF is able to act as a transcriptional repressor, we tested FAZF by coexpressing the protein along with a reporter gene linked to a promoter containing multimerized IL3R-PLZF target sites. We found that both PLZF and FAZF specifically repressed expression of this reporter gene, but not the parental tk-luciferase reporter (Fig 8A). The transcriptonal repression was dependent on the amount of plasmid transfected, and when both PLZF and FAZF were expressed, there was an additive effect on repression (Fig8B). Taken together, the binding and repression data suggest that not only do PLZF and FAZF proteins bind to the same DNA sequence, but they repress transcription by a similar mechanism.

Fig. 8.

FAZF is a transcriptional repressor. (A) 293T cells were cotransfected in quadruplicate with a luciferase reporter gene containing four copies of a high-affinity PLZF binding site (0.15 μg) from the IL-3–receptor promoter linked 5′ to the HSV-tk promoter or the parental tk-luciferase reporter (0.15 μg) along with 0.85 μg of expression vectors for PLZF or FAZF as indicated or the pSG5 expression vector. At 48 hours after transfection, the cells were assayed for luciferase activity. (B) Increasing amounts of the PLZF or FAZF expression vector or a combination of both vectors was cotransfected in quadruplicate along with the IL3-tk-luciferase promoter (0.1 μg). Luciferase activity was assayed as above.

Fig. 8.

FAZF is a transcriptional repressor. (A) 293T cells were cotransfected in quadruplicate with a luciferase reporter gene containing four copies of a high-affinity PLZF binding site (0.15 μg) from the IL-3–receptor promoter linked 5′ to the HSV-tk promoter or the parental tk-luciferase reporter (0.15 μg) along with 0.85 μg of expression vectors for PLZF or FAZF as indicated or the pSG5 expression vector. At 48 hours after transfection, the cells were assayed for luciferase activity. (B) Increasing amounts of the PLZF or FAZF expression vector or a combination of both vectors was cotransfected in quadruplicate along with the IL3-tk-luciferase promoter (0.1 μg). Luciferase activity was assayed as above.

Close modal

We report here that FANCC interacts with a novel protein, FAZF, which we show functions as a transcriptional repressor. The FAZF/FANCC interaction maps to a central region in FANCC, which is deleted in patients with a severe FA phenotype.14,53 Moreover, we found that wild-type FANCC, but not the L554P mutant protein, can colocalize with FAZF in nuclear foci. Thus, disturbing the FANCC/FAZF interaction may play a role in FA.

The FAZF protein is similar to transcriptional repressors of the BTB/POZ subfamily of Krüppel-like zinc finger proteins (reviewed in Albagli et al25 and Bardwell et al26), which have been shown to be important for development, tissue-specific proliferation and differentiation, and neoplasia. In particular, FAZF is similar to the PLZF gene, which is translocated and fused to the RARα locus t(11;17)(q23;q21) in a subset of patients with APL.27,29,35 Recently, the mechanism of transcriptional repression of PLZF has been shown to occur through binding of specific promoter sequences61,62 followed by recruitment of nuclear receptor Sin-3/histone deactylase corepressor complexes.30-32,70-72 Like PLZF, we found that FAZF is able to repress transcription, suggesting that the function of these two proteins is, at least in part, overlapping.

The BTB/POZ protein-protein interaction motif is a highly conserved domain found in a number of eukaryotic proteins involved in cancer including the PLZF protein,29 BCL-6,37 and HIC (hypermethylated in cancer).73 The BTB/POZ domain mediates self-association26,59,60 and specific heterodimeric association with other BTB/POZ proteins (eg, PLZF,74 LRF, and LACZ-3/BCL-641). Recent evidence suggests that even though BTB/POZ domains may have significant homology and are able to interact with a related set of corepressor components, they may differ in their interactions with the corepressor complex.58,75Thus, crosstalk through heterodimer formation combined with the subtle differences in corepressor interactions suggests that transcriptional regulation by the proteins in this family is highly permuted. In this regard, we found that the FANCC/FAZF interaction is apparently not dependent on the BTB/POZ domain of FAZF, potentially allowing this region to form homodimeric and heterodimeric complexes with other proteins. Indeed, we report here that FAZF forms homodimers and heterodimerizes with PLZF. It will be interesting to determine if unique binding partners exist for the FAZF POZ domain.

The FANCC/FAZF interaction suggests that FANCC may play a role in the repression of gene expression, potentially in response to DNA damage. The nature of the target genes is unknown. The only PLZF target gene reported so far is cyclin A2. Enforced expression of PLZF delayed the transit of cells though the G1 and S phases of the cell cycle, an effect that was reversed by ectopic expression of cyclin A in these cells.76,77 The cyclin A2 promoter contains two PLZF binding sites that are required for the protein to repress this promoter. It is plausible that FAZF could have a similar effect on cyclin A expression and the cell cycle, perhaps reflecting a role in inhibiting cell growth in response to DNA damage. Although the FAZF protein can bind to the same binding site as PLZF, the fact that the FAZF protein only contains three zinc fingers whereas PLZF contains nine motifs suggests that the range of target genes bound by these two proteins may only partially overlap. Future studies will determine the effects of FAZF in cell growth control and DNA damage response.

Immunofluoresence analysis of FANCC in this study indicated that FANCC is expressed in both cytoplasmic and nuclear compartments, as reported previously.16-19 However, L554P was not found in the nuclear compartment at wild-type levels. Failure, or reduced ability, of L554P to enter the nucleus could account for its defect. Confirmation awaits comparison of endogenous wild-type and L554P localization that is currently not possible because of low endogenous expression levels. We also observed that a subset of FANCC-expressing cells contained nuclear foci, and suggest that this is a feature of wild-type FANCC localization. The following evidence suggests that these foci might be functionally significant. First, although expressed at comparable levels, the signal observed for the L554P mutant protein was greatly reduced in the nucleus as suggested by earlier studies,19 and was consequently unable to form nuclear foci. That L554P could be highly expressed in the cytoplasm and not “forced” into the nucleus also argues against the notion that nuclear localization and nuclear foci are simply an artifact of overexpression. Second, FANCA has also been observed in nuclear foci.68 Third, D’Andrea et al18,78 have reported that the nuclear fractions of FANCA and FANCC, but not L554P, form a complex and that this nuclear complex is functional. Taken together, these observations support the notion that foci formation may be biologically significant. Finally, we determined that FAZF, like other members of its family, is located primarily in nuclear foci.26,40,47,60,67 Our coimmunoprecipitation and colocalization experiments independently support the FAZF/FANCC interaction detected by two-hybrid analysis and in vitro binding. We found that in some cells, colocalization of FANCC and FAZF foci was extensive, whereas only partial overlap was observed in other cells, suggesting that the FAZF/FANCC interaction may be transient or conditional. This pattern of partially overlapping foci has been described for a series of colocalized proteins including BRCA1/hRad51,79 Arenavirus proteins/PML,80retinoblastoma protein/PML,81 and PML/PLZF.74Given these complexities, and the fact that the FA pathway is novel, further studies will be required to understand the consequences of the FAZF/FANCC interaction and the potential role of transcriptional repression in the genomic instability observed in FA.

The authors are grateful to their colleagues Mike Forte and Beth Blanchly-Dyson for initial assistance with the two-hybrid studies. The authors thank the members of the Fanconi anemia research community for stimulating discussions, in particular Hans Joenje, Mike Heinrich, Johan de Winter, Quinten Waisfisz, and Henri van de Vrugt for their interest and advice.

Supported by NIH grants HL56045 (M.E.H.), CA59938 (J.D.L., A.Z.), and K08 CA73762 (A.M.); the Medical Research Foundation of Oregon and the Fanconi Anemia Research Fund (M.E.H.); and the American Cancer Society DHP160 (J.D.L.). J.D.L. is a Scholar of the Leukemia Society of America.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Fanconi
G
Familial constitutional panmyelocytopathy, Fanconi’s anemia (F.A.). I. Clinical aspects.
Semin Hematol
4
1967
233
2
Butturini
A
Gale
RP
Verlander
PC
Adler-Brecher
B
Gillio
AP
Auerbach
AD
Hematologic abnormalities in Fanconi anemia: An International Fanconi Anemia Registry study.
Blood
84
1994
1650
3
Strathdee
CA
Buchwald
M
Molecular and cellular biology of Fanconi anemia.
Am J Pediatr Hematol Oncol
14
1992
177
4
Auerbach
A
Buchwald
M
Joenje
H
Fanconi Anemia
The Genetic Basis of Human Cancer.
Vogelstein
B
Kinzler
KW
1997
McGraw Hill
New York, NY
5
Liu
JM
Buchwald
M
Walsh
CE
Young
NS
Fanconi anemia and novel strategies for therapy.
Blood
84
1994
3995
6
Dutrillaux
B
Aurias
A
Dutrillaux
AM
Buriot
D
Prieur
M
The cell cycle of lymphocytes in Fanconi anemia.
Hum Genet
62
1982
327
7
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
8
Kinzler
KW
Vogelstein
B
Cancer-susceptibility genes. Gatekeepers and caretakers.
Nature
386
1997
761
9
Joenje
H
Oostra
AB
Wijker
M
di Summa
FM
van Berkel
CG
Rooimans
MA
Ebell
W
van Weel
M
Pronk
JC
Buchwald
M
Arwert
F
Evidence for at least eight Fanconi anemia genes.
Am J Hum Genet
61
1997
940
10
Buchwald
M
Complementation groups: One or more per gene?
Nat Genet
11
1995
228
11
Strathdee
CA
Gavish
H
Shannon
WR
Buchwald
M
Cloning of cDNAs for Fanconi’s anaemia by functional complementation.
Nature
356
1992
763
12
de Winter
JP
Waisfisz
Q
Rooimans
MA
van Berkel
CG
Bosnoyan-Collins
L
Alon
N
Carreau
M
Bender
O
Demuth
I
Schindler
D
Pronk
JC
Arwert
F
Hoehn
H
Digweed
M
Buchwald
M
Joenje
H
The Fanconi anaemia group G gene FANCG is identical with XRCC9.
Nat Genet
20
1998
281
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
CG
Strunk
MH
Gille
JJ
Pals
G
Kruyt
FA
Pronk
JC
Arwert
F
Buchwald
M
Joenje
H
Expression cloning of a cDNA for the major Fanconi anaemia gene, FAA.
Nat Genet
14
1996
320
14
Whitney
MA
Saito
H
Jakobs
PM
Gibson
RA
Moses
RE
Grompe
M
A common mutation in the FACC gene causes Fanconi anaemia in Ashkenazi Jews.
Nat Genet
4
1993
202
15
Waisfisz
Q
Saar
K
Morgan
NV
Altay
C
Leegwater
PA
de Winter
JP
Komatsu
K
Evans
GR
Wegner
RD
Reis
A
Joenje
H
Arwert
F
Mathew
CG
Pronk
JC
Digweed
M
The Fanconi Anemia Group E gene, FANCE, maps to chromosome 6p.
Am J Hum Genet
64
1999
1400
16
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
17
Youssoufian
H
Localization of Fanconi anemia C protein to the cytoplasm of mammalian cells.
Proc Natl Acad Sci USA
91
1994
7975
18
Kupfer
GM
Naf
D
Suliman
A
Pulsipher
M
D’Andrea
AD
The Fanconi anaemia proteins, FAA and FAC, interact to form a nuclear complex.
Nat Genet
17
1997
487
19
Hoatlin
ME
Christianson
TA
Keeble
WW
Hammond
AT
Zhi
Y
Heinrich
MC
Tower
PA
Bagby
GC
Jr
The Fanconi anemia group C gene product is located in both the nucleus and cytoplasm of human cells.
Blood
91
1998
1418
20
Kupfer
G
Yamashita
T
Naf
D
Suliman
A
Asano
S
D’Andrea
A
The Fanconi anemia polypeptide, FAC, binds to the cyclin-dependent kinase, cdc2.
Blood
90
1997
1047
21
Hoshino
T
Wang
J
Devetten
MP
Iwata
N
Kajigaya
S
Wise
RJ
Liu
JM
Youssoufian
H
Molecular chaperone GRP94 binds to the Fanconi anemia group C protein and regulates its intracellular expression.
Blood
91
1998
4379
22
Kruyt
FA
Hoshino
T
Liu
JM
Joseph
P
Jaiswal
AK
Youssoufian
H
Abnormal microsomal detoxification implicated in Fanconi anemia group C by interaction of the FAC protein with NADPH cytochrome P450 reductase.
Blood
92
1998
3050
23
Garcia-Higuera
I
Kuang
Y
Naf
D
Wasik
J
D’Andrea
AD
Fanconi Anemia Proteins FANCA, FANCC, and FANCG/XRCC9 interact in a Functional Nuclear Complex.
Mol Cell Biol
19
1999
4866
24
Waisfisz
Q
de Winter
JP
Kruyt
F
de Groot
J
van der Weel
L
Dijkmans
L
Zhi
Y
Arwert
F
Scheper
R
Youssoufian
H
Hoatlin
M
Joenje
H
Evidence for a physical complex of the Fanconi anemia proteins FANCG/XRCC9 and FANCA.
Proc Natl Acad Sci USA
96
1999
10320
25
Albagli
O
Dhordain
P
Deweindt
C
Lecocq
G
Leprince
D
The BTB/POZ domain: A new protein-protein interaction motif common to DNA-and actin-binding proteins.
Cell Growth Differ
6
1995
1193
26
Bardwell
VJ
Treisman
R
The POZ domain: A conserved protein-protein interaction motif.
Genes Dev
8
1994
1664
27
Melnick
A
Licht
J
Deconstructing a disease: RAR α, its fusion proteins and their roles in the pathogenesis of acute promyelocytic leukemia.
Blood
93
1999
3167
28
Hong
SH
David
G
Wong
CW
Dejean
A
Privalsky
ML
SMRT corepressor interacts with PLZF and with the PML-retinoic acid receptor alpha (RARalpha) and PLZF-RARalpha oncoproteins associated with acute promyelocytic leukemia.
Proc Natl Acad Sci USA
94
1997
9028
29
Chen
Z
Brand
NJ
Chen
A
Chen
SJ
Tong
JH
Wang
ZY
Waxman
S
Zelent
A
Fusion between a novel Kruppel-like zinc finger gene and the retinoic acid receptor-alpha locus due to a variant t(11;17) translocation associated with acute promyelocytic leukaemia.
EMBO J
12
1993
1161
30
He
LZ
Guidez
F
Tribioli
C
Peruzzi
D
Ruthardt
M
Zelent
A
Pandolfi
PP
Distinct interactions of PML-RARalpha and PLZF-RARalpha with co-repressors determine differential responses to RA in APL.
Nat Genet
18
1998
126
31
David
G
Alland
L
Hong
SH
Wong
CW
DePinho
RA
Dejean
A
Histone deacetylase associated with mSin3A mediates repression by the acute promyelocytic leukemia-associated PLZF protein.
Oncogene
16
1998
2549
32
Guidez
F
Ivins
S
Zhu
J
Soderstrom
M
Waxman
S
Zelent
A
Reduced retinoic acid-sensitivities of nuclear receptor corepressor binding to PML- and PLZF-RARalpha underlie molecular pathogenesis and treatment of acute promyelocytic leukemia.
Blood
91
1998
2634
33
Baron
BW
Nucifora
G
McCabe
N
Espinosa
R
3d
Le Beau
MM
McKeithan
TW
Identification of the gene associated with the recurring chromosomal translocations t(3;14)(q27;q32) and t(3;22)(q27;q11) in B-cell lymphomas.
Proc Natl Acad Sci USA
90
1993
5262
34
Chen
SJ
Zelent
A
Tong
JH
Yu
HQ
Wang
ZY
Derre
J
Berger
R
Waxman
S
Chen
Z
Rearrangements of the retinoic acid receptor alpha and promyelocytic leukemia zinc finger genes resulting from t(11;17)(q23;q21) in a patient with acute promyelocytic leukemia.
J Clin Invest
91
1993
2260
35
Licht
JD
Chomienne
C
Goy
A
Chen
A
Scott
AA
Head
DR
Michaux
JL
Wu
Y
DeBlasio
A
Miller
WH
Jr
Zelenetz
AD
Wilman
CL
Chen
Z
Chen
S-J
Zelent
A
Macintyre
E
Veil
A
Cortes
J
Kantarjian
H
Waxman
S
Clinical and molecular characterization of a rare syndrome of acute promyelocytic leukemia associated with translocation (11;17).
Blood
85
1995
1083
36
Miki
T
Kawamata
N
Arai
A
Ohashi
K
Nakamura
Y
Kato
A
Hirosawa
S
Aoki
N
Molecular cloning of the breakpoint for 3q27 translocation in B-cell lymphomas and leukemias.
Blood
83
1994
217
37
Ye
BH
Lista
F
Lo Coco
F
Knowles
DM
Offit
K
Chaganti
RS
Dalla-Favera
R
Alterations of a zinc finger-encoding gene, BCL-6, in diffuse large-cell lymphoma.
Science
262
1993
747
38
Dent
AL
Shaffer
AL
Yu
X
Allman
D
Staudt
LM
Control of inflammation, cytokine expression, and germinal center formation by BCL-6.
Science
276
1997
589
39
Ye
BH
Cattoretti
G
Shen
Q
Zhang
J
Hawe
N
de Waard
R
Leung
C
Nouri-Shirazi
M
Orazi
A
Chaganti
RS
Rothman
P
Stall
AM
Pandolfi
PP
Dalla-Favera
R
The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation.
Nat Genet
16
1997
161
40
Reid
A
Gould
A
Brand
N
Cook
M
Strutt
P
Li
J
Licht
J
Waxman
S
Krumlauf
R
Zelent
A
Leukemia translocation gene, PLZF, is expressed with a speckled nuclear pattern in early hematopoietic progenitors.
Blood
86
1995
4544
41
Davies
JM
Hawe
N
Kabarowski
J
Huang
QH
Zhu
J
Brand
NJ
Leprince
D
Dhordain
P
Cook
M
Morriss-Kay
G
Zelent
A
Novel BTB/POZ domain zinc-finger protein, LRF, is a potential target of the LAZ-3/BCL-6 oncogene.
Oncogene
18
1999
365
42
Grimm
C
Sporle
R
Schmid
TE
Adler
ID
Adamski
J
Schughart
K
Graw
J
Isolation and embryonic expression of the novel mouse gene Hic1, the homologue of HIC1, a candidate gene for the Miller-Dieker syndrome.
Hum Mol Genet
8
1999
697
43
Hawe
N
Soares
V
Niswander
L
Cattoretti
G
Pandolfi
P
Targeted disruption of the PLZF gene of acute promyelocytic leukemia results in phocomelia and altered spermatogenesis.
Blood
88
1996
1151
(abstr)
44
Oi
VT
Herzenberg
L
Immunoglobulin-producing hybrid cell lines
Selected Methods in Cellular Immunology.
Mishell
B
Shiigi
SM
1980
351
W. H. Freeman and Company
New York, NY
45
Durfee
T
Becherer
K
Chen
P
Yeh
P
Yang
Y
Kilburn
A
Lee
W
Elledge
S
The retinoblastoma protein associates with the protein phosphatase type 1 catalytic subunit.
Genes Dev
7
1993
555
46
James
P
Halladay
J
Craig
EA
Genomic libraries and a host strain designed for highly efficient two-hybrid selection in yeast.
Genetics
144
1996
1425
47
Licht
JD
Shaknovich
R
English
MA
Melnick
A
Li
JY
Reddy
JC
Dong
S
Chen
SJ
Zelent
A
Waxman
S
Reduced and altered DNA-binding and transcriptional properties of the PLZF-retinoic acid receptor-alpha chimera generated in t(11;17)-associated acute promyelocytic leukemia.
Oncogene
12
1996
323
48
Statview Reference Software.
1998
SAS Institute
Cary, NC
49
Ausubel
FM
Brent
R
Kingston
RE
Moore
DD
Seidman
JG
Smith
JA
Struhl
K
Current Protocols in Molecular Biology.
1989
Wiley
New York, NY
50
Andrews
NC
Faller
DV
A rapid micropreparation technique for extraction of DNA-binding proteins from limiting numbers of mammalian cells.
Nucleic Acids Res
19
1991
2499
51
Li
X
Lopez-Guisa
JM
Ninan
N
Weiner
EJ
Rauscher
FJ
3rd
Marmorstein
R
Overexpression, purification, characterization, and crystallization of the BTB/POZ domain from the PLZF oncoprotein.
J Biol Chem
272
1997
27324
51a
Ball
H
Melnick
A
Shaknovich
R
Kohanski
R
Licht
JD
The promyelocytic leukemia zinc finger (PLZF) protein binds DNA in a high molecular weight complex associated with cdc2 kinase.
Nucleic Acids Res
27
1999
4106
52
Buchwald
M
Moustacchi
E
Is Fanconi anemia caused by a defect in the processing of DNA damage?
Mutat Res
408
1998
75
53
Gillio
AP
Verlander
PC
Batish
SD
Giampietro
PF
Auerbach
AD
Phenotypic consequences of mutations in the Fanconi anemia FAC gene: An International Fanconi Anemia Registry study.
Blood
90
1997
105
54
Whitney
MA
Jakobs
P
Kaback
M
Moses
RE
Grompe
M
The Ashkenazi Jewish Fanconi anemia mutation: Incidence among patients and carrier frequency in the at-risk population.
Hum Mutat
3
1994
339
55
Altschul
SF
Madden
TL
Schaffer
AA
Zhang
J
Zhang
Z
Miller
W
Lipman
DJ
Gapped BLAST and PSI-BLAST: A new generation of protein database search programs.
Nucleic Acids Res
25
1997
3389
56
Altschul
SF
Gish
W
Miller
W
Myers
EW
Lipman
DJ
Basic local alignment search tool.
J Mol Biol
215
1990
403
57
Zollman
S
Godt
D
Prive
G
Couder
JL
Laski
F
The BTB domain, found primarily in zinc finger proteins, defines an evolutionarily conserved family that includes several developmentally regulated genes in Drosophila.
Proc Natl Acad Sci USA
91
1994
10717
58
Daniel
JM
Reynolds
AB
The catenin p120(ctn) interacts with kaiso, a novel BTB/POZ domain zinc finger transcription factor.
Mol Cell Biol
19
1999
3614
59
Ahmad
KF
Engel
CK
Prive
GG
Crystal structure of the BTB domain from PLZF.
Proc Natl Acad Sci USA
95
1998
12123
60
Dong
S
Zhu
J
Reid
A
Strutt
P
Guidez
F
Zhong
HJ
Wang
ZY
Licht
J
Waxman
S
Chomienne
C
Chen
Z
Zelent
A
Chen
S-J
Amino-terminal protein-protein interaction motif (POZ-domain) is responsible for activities of the promyelocytic leukemia zinc finger-retinoic acid receptor-alpha fusion protein.
Proc Natl Acad Sci USA
93
1996
3624
61
Li
JY
English
MA
Ball
HJ
Yeyati
PL
Waxman
S
Licht
JD
Sequence-specific DNA binding and transcriptional regulation by the promyelocytic leukemia zinc finger protein.
J Biol Chem
272
1997
22447
62
Sitterlin
D
Tiollais
P
Transy
C
The RAR alpha-PLZF chimera associated with Acute Promyelocytic Leukemia has retained a sequence-specific DNA-binding domain.
Oncogene
14
1997
1067
63
Ziemin-van der Poel
S
McCabe
NR
Gill
HJ
Espinosa
R
III
Patel
Y
Harden
A
Rubinelli
P
Smith
SD
LeBeau
MM
Rowley
JD
Diaz
MO
Identification of a gene, MLL, that spans the breakpoint in 11q23 translocations associated with human leukemias.
Proc Natl Acad Sci USA
88
1991
10735
64
Gu
Y
Nakamura
T
Alder
H
Prasad
R
Canaani
O
Cimino
G
Croce
CM
Canaani
E
The t(4;11) chromosome translocation of human acute leukemias fuses the ALL-1 gene, related to Drosophila trithorax, to the AF-4 gene.
Cell
71
1992
701
65
Tkachuk
DC
Kohler
S
Cleary
ML
Involvement of a homolog of Drosophila trithorax by 11q23 chromosomal translocations in acute leukemias.
Cell
71
1992
691
66
Djabali
M
Selleri
L
Parry
P
Bower
M
Young
BD
Evans
GA
A trithorax-like gene is interrupted by chromosome 11q23 translocations in acute leukaemias [published erratum appears in Nat Genet 1993 Aug;4(4):431].
Nat Genet
2
1992
113
67
Dhordain
P
Albagli
O
Ansieau
S
Koken
MH
Deweindt
C
Quief
S
Lantoine
D
Leutz
A
Kerckaert
JP
Leprince
D
The BTB/POZ domain targets the LAZ3/BCL6 oncoprotein to nuclear dots and mediates homomerisation in vivo.
Oncogene
11
1995
2689
68
Naf
D
Kupfer
GM
Suliman
A
Lambert
K
D’Andrea
AD
Functional activity of the fanconi anemia protein FAA requires FAC binding and nuclear localization.
Mol Cell Biol
18
1998
5952
69
Savoia
A
Garcia-Higuera
I
D’Andrea
AD
Nuclear localization of the Fanconi anemia protein FANCC is required for functional activity [letter].
Blood
93
1999
4025
70
Hong
SH
Wong
CW
Privalsky
ML
Signaling by tyrosine kinases negatively regulates the interaction between transcription factors and SMRT (silencing mediator of retinoic acid and thyroid hormone receptor) corepressor.
Mol Endocrinol
12
1998
1161
71
Wong
CW
Privalsky
ML
Components of the SMRT corepressor complex exhibit distinctive interactions with the POZ domain oncoproteins PLZF, PLZF-RARalpha, and BCL-6.
J Biol Chem
273
1998
27695
72
Wong
CW
Privalsky
ML
Transcriptional repression by the SMRT-mSin3 corepressor: Multiple interactions, multiple mechanisms, and a potential role for TFIIB.
Mol Cell Biol
18
1998
5500
73
Wales
MM
Biel
MA
el Deiry
W
Nelkin
BD
Issa
JP
Cavenee
WK
Kuerbitz
SJ
Baylin
SB
p53 activates expression of HIC-1, a new candidate tumour suppressor gene on 17p13.3.
Nat Med
1
1995
570
74
Koken
MH
Reid
A
Quignon
F
Chelbi-Alix
MK
Davies
JM
Kabarowski
JH
Zhu
J
Dong
S
Chen
S
Chen
Z
Tan
CC
Licht
J
Waxman
S
de The
H
Zelent
A
Leukemia-associated retinoic acid receptor alpha fusion partners, PML and PLZF, heterodimerize and colocalize to nuclear bodies.
Proc Natl Acad Sci USA
94
1997
10255
75
Hong
SH
Privalsky
ML
Retinoid isomers differ in the ability to induce release of SMRT corepressor from retinoic acid receptor-alpha.
J Biol Chem
274
1999
2885
76
Shaknovich
R
Yeyati
PL
Ivins
S
Melnick
A
Lempert
C
Waxman
S
Zelent
A
Licht
JD
The promyelocytic leukemia zinc finger protein affects myeloid cell growth, differentiation, and apoptosis.
Mol Cell Biol
18
1998
5533
77
Yeyati
PL
Shaknovich
R
Boterashvili
S
Li
J
Ball
HJ
Waxman
S
Nason-Burchenal
K
Dmitrovsky
E
Zelent
A
Licht
JD
Leukemia translocation protein PLZF inhibits cell growth and expression of cyclin A.
Oncogene
18
1999
925
78
Kupfer
G
Naf
D
Garcia-Higuera
I
Wasik
J
Cheng
A
Yamashita
T
Tipping
A
Morgan
N
Mathew
CG
D’Andrea
AD
A patient-derived mutant form of the Fanconi anemia protein, FANCA, is defective in nuclear accumulation.
Exp Hematol
27
1999
587
79
Scully
R
Chen
J
Plug
A
Xiao
Y
Weaver
D
Feunteun
J
Ashley
T
Livingston
DM
Association of BRCA1 with Rad51 in mitotic and meiotic cells.
Cell
88
1997
265
80
Borden
KL
Campbelldwyer
EJ
Carlile
GW
Djavani
M
Salvato
MS
Two RING finger proteins, the oncoprotein PML and the arenavirus Z protein, colocalize with the nuclear fraction of the ribosomal P proteins.
J Virol
72
1998
3819
81
Alcalay
M
Tomassoni
L
Colombo
E
Stoldt
S
Grignani
F
Fagioli
M
Szekely
L
Helin
K
Pelicci
PG
The promyelocytic leukemia gene product (PML) forms stable complexes with the retinoblastoma protein.
Mol Cell Biol
18
1998
1084
82
Lo ten Foe
JR
Barel
MT
Thuss
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

Author notes

Address reprint requests to Maureen E. Hoatlin, PhD, Division of Hematology and Medical Oncology, Oregon Health Sciences University, 3181 SW Sam Jackson Park Rd, Portland, OR 97201; e-mail:hoatlinm@OHSU.edu.

Sign in via your Institution