Acute promyelocytic leukemia (APL) is typified by the reciprocal translocation, t(15; 17)(q22; q21), leading to the formation of PML-RARα and RARα-PML fusion genes. We have characterized 7 cases of morphologic APL found to lack the t(15; 17) on conventional cytogenetic assessment. In 6 of 7 cases, cryptic PML-RARα rearrangements were identified by reverse transcriptase-polymerase chain reaction and fluorescent in situ hybridization (FISH); whereas, in the remaining patient, APL was associated with the variant translocation, t(11; 17)(q23; q12-21), leading to the formation of PLZF-RARα and RARα-PLZF fusion genes. In each of the cases with cryptic PML-RARα rearrangements, PML-RARα transcripts were detected in the absence of RARα-PML, consistent with the concept that PML-RARα is the critical oncogenic fusion protein. In 4 of these cases with evaluable metaphase spreads, the occurrence of a nonreciprocal translocation was confirmed by FISH with sole formation of the PML-RARα fusion gene; in 3 cases with morphologically normal chromosomes 15 and 17, RARα was inserted into PML on 15q, whereas in the remaining patient the PML-RARα fusion arose due to insertion of 15q-derived material including PML into RARα on 17q. Immunofluorescence studies were performed using antibodies raised against PML and PIC 1, a ubiquitin-homology domain protein previously identified as an interaction partner of PML. In acute myeloid leukemia (AML) of subtypes other than M3, PIC 1 was localized to the nuclear membrane and colocalized with PML within discrete nuclear bodies. In APL cases with cryptic PML-RARα rearrangements, the characteristic microparticulate pattern of PML staining was detected with partial colocalization with PIC 1, indicative of disruption of the nuclear bodies; whereas in t(11; 17)-associated APL, PML and PIC 1 remained colocalized within discrete nuclear bodies, as observed in non-APL cases. Although deregulation of the putative growth suppressor PML and delocalization of other nuclear body constituents have been advocated to play a key role in the development of t(15; 17)-associated APL, the present study shows that disruption of PML nuclear bodies per se is not a prerequisite for the pathogenesis of APL.

ACUTE PROMYELOCYTIC leukemia (APL) is typified by the reciprocal translocation, t(15; 17)(q22; q21),1 leading to the formation of PML-RARα and RARα-PML fusion genes (reviewed by Grimwade and Solomon2 and references therein). PML-RARα, transcribed from add(15q), retains virtually all the domains considered to be of functional importance to both PML and RARα and has therefore traditionally been considered to play a key role in leukemogenesis, which has recently been confirmed using a transgenic model.3,4 Any role for del(17q)-derived RARα-PML in the development of APL remains unclear, particularly because reciprocal transcripts are detected in only 81% of patients5; nevertheless, a case of APL apparently lacking a PML-RARα fusion gene and in which only RARα-PML transcripts could be detected has recently been reported.6 The molecular pathogenesis of APL is believed to reflect two key processes: leukemic transformation coupled with a block in myeloid differentiation such that the marrow becomes replaced by abnormal promyelocytes.7 In APL cases associated with a PML-RARα rearrangement, this differentiation block may be overcome by retinoids such as all-trans retinoic acid (ATRA).8 To understand these phenomena, much effort has been devoted to the study of the physiologic roles of RARα and PML.

RARα is a member of the steroid hormone nuclear receptor family, serving as a transcription factor mediating the effect of retinoic acid at specific response elements (reviewed by Stunnenberg9 ). In common with vitamin D and thyroid hormone receptors, high-affinity DNA binding of retinoic acid receptors (RARs) requires heterodimerization with a member of the retinoid X receptor family.10,11 Integrity of these retinoid signaling pathways is critical for normal embryogenesis (reviewed by Grimwade and Solomon2 and references therein) and postnatal myeloid differentiation.12 13 

In contrast to RARα, the role of PML remains less clearly defined. Initial claims that it also serves as a transcription factor on the basis of N-terminal zinc-binding RING finger and B-box domains14,15 remain unsubstantiated, and indeed more recent work has failed to demonstrate specific DNA-binding for the majority of RING-family members (reviewed by Saurin et al16 ). With the development of appropriate antisera, PML has been found to be predominantly localized to the nucleus within structures known as PML nuclear bodies.17-20 These are composed of several proteins of unknown function, including NDP52,21 Sp100, and Sp140, which were identified as targets for autoantibodies in patients with primary biliary cirrhosis,22,23 and PIC 1, a ubiquitin-homology domain protein, which has been found to interact directly with PML.24 A variety of experimental approaches have implicated PML in immunologic responses (Grimwade and Solomon2 and references therein) and there is some evidence to suggest that PML itself or components of the nuclear bodies are cell-cycle regulated and can mediate growth-suppressor activity.25-28 Cells from a wide range of tissues and blasts from leukemic subtypes other than APL typically demonstrate 10 to 30 discrete nuclear bodies when stained with PML antisera. Whereas, in APL cases associated with the t(15; 17), a microparticulate pattern of PML staining is characteristic,17-19 reflecting disruption of nuclear bodies due to an interaction between PML and PML-RARα.29 This process may promote leukemogenesis by delocalizing the putative growth-suppressor PML and other nuclear body components; this, coupled with an abnormal pattern of retinoid responses also mediated by the fusion protein, compounded by RXR sequestration could account for the block in myeloid differentiation that characterizes the disease. Treatment of such cases with ATRA leads to complete remission by terminal differentiation of the leukemic clone associated with release of inhibitory effects of PML-RARα at retinoid response elements14,30-32 and degradation of the fusion protein33,34 accompanied by normalization of nuclear body architecture.17-20 Rare cases of PML-RARα–mediated APL have been identified that fail to respond to retinoids associated with persistence of the microparticulate PML nuclear staining pattern.19,35 On the basis of these findings it has been suggested that disruption of PML nuclear bodies is critical to the pathogenesis of APL; furthermore, it has been advocated that reconstitution of normal nuclear architecture is essential to permit differentiation in the presence of retinoids.19 

Although ATRA therapy is unable to sustain long-term remission in APL,36-38 recent studies have shown that a combined treatment approach using ATRA and chemotherapy confers significant improvements in disease-free survival compared with chemotherapy alone.39,40 Because a favorable response to ATRA appears to be restricted to patients with the t(15; 17),8,41 establishing the presence of this cytogenetic change, or in its absence identification of a PML-RARα rearrangement, is fundamental for optimal treatment of patients and meaningful analysis of APL trials. Although early studies in specialized centers reported the presence of the t(15; 17) in all cases of APL42; it is now clear that some cases with morphologic acute myeloid leukemia (AML) M3 reflect cryptic PML-RARα rearrangements,6,43,44 whereas in others RARα is fused to a partner other than PML. Thus far, three such alternative fusion partners have been identified, including the novel Krüppel-like zinc finger protein PLZF involved in the t(11; 17)(q23; q21)45; nucleophosmin, an RNA processing protein disrupted by the t(5; 17)(q32; q12)46; and most recently NuMA, which is involved in control of mitosis and is rearranged by the t(11; 17)(q13; q21).47 Although such variant translocations are extremely rare, accounting for less than 1% of morphologic APL,48 elucidation of the mechanisms underlying leukemogenesis in these cases is likely to provide considerable insight into the processes involved in the development of PML-RARα–mediated disease and in particular as to whether disruption of PML nuclear bodies is fundamental to the pathogenesis of APL. Cases with variant fusion translocations also afford the opportunity to dissect out mechanisms leading to leukemic transformation from those mediating the block in myeloid differentiation and its reversal by retinoids. In the present study, we have characterized 7 cases of morphologic APL found to lack the t(15; 17) on conventional cytogenetic assessment and considered their implications for the pathogenesis of APL as a whole.

Patients and cytogenetics. This study considers 7 patients with morphologic APL who were found to lack the t(15; 17) on conventional cytogenetic assessment. Five patients were drawn from the MRC ATRA trial; details of the treatment protocol have been described elsewhere.5 Cytogenetic assessment was undertaken at local centers or by the central UK MRC AML trials cytogenetics service at University College Hospital, London, according to standard methods.49 In each case in which APL was associated with a normal karyotype, preparations were subject to at least 24 hours of culture and a minimum of 20 metaphases were examined.

Reverse transcriptase-polymerase chain reaction (RT-PCR) and sequence analysis. Details of bone marrow and peripheral blood sample preparation, RNA extraction, and RT-PCR protocols to detect PML-RARα and RARα 1-PML fusion transcripts in conjunction with RARα and PML as controls for RNA integrity have been fully described elsewhere.5 50 Using cDNA generated by the same method, PLZF-RARα and RARα-PLZF transcripts were amplified using nested PCR in material derived from a patient with t(11; 17)(q23; q12-21) identified by cytogenetics. Primers used for PLZF-RARα PCR were as follows: PLZF external, 5′-TCCAGAGGGAGCTGTTCAGC-3′; RARα external, 5′-TCTTCTGGATGCTGCGGCGG-3′; PLZF internal, 5′-TCGAGCTTCCTGATAACGAG-3′; and RARα internal, 5′-GGCGCTGACCCCATAGTGGT-3′. Primers for RARα 1-PLZF nested PCR comprised the following: RARα external, 5′-GGCCAGCAACAGCAGCTCCT-3′; PLZF external, 5′-ATGTCAGTGCCAGTATGGGT-3′; RARα internal, 5′-GGTGCCTCCCTACGCCTTCT-3′; and PLZF internal, 5′-CACTGATCACAGACAAAGGC-3′.

PCR was performed in a 50 μL reaction, with 1 μL of the first round PCR products used as template for the second round of PCR, as previously described.51 First and second round PCR reactions comprised 35 cycles, each consisting of 1 minute of denaturation at 95°C, 1 minute of annealing at 57°C, and 1 minute of extension at 72°C, followed by 10 minutes of extension at 72°C (OmniGene apparatus; Hybaid, Teddington, Middlesex, UK). PCR products were size-separated on ethidium bromide-stained 1.5% agarose gels, as previously described.50Bcr 1 (intron 6) and bcr 2 (exon 6) PML breakpoints were distinguished by sequence analysis of PML-RARα PCR products using previously described methods and primers.5PLZF-RARα and RARα-PLZF PCR products were similarly sequenced, using appropriate internal primers, with an automated sequencer (377; ABI, Perkin-Elmer, CA).

Fluorescent in situ hybridization (FISH). FISH using ICRF PML and RARα cosmid probes (15.5 and 121, respectively) was performed to detect PML-RARα fusion genes in patients lacking the t(15; 17), using previously described methods.43 PML cosmid 15.5 encompasses the 5′ region of the gene, including exons 1 and 2, whereas RARα cosmid 121 spans the APL breakpoint region on chromosome 17.43 In APL cases associated with the t(15; 17), a dual signal is detected with these probes on the derivative chromosome 15, identifying the site of the PML-RARα fusion, in addition to single hybridization signals corresponding to the normal PML and RARα loci on 15q and 17q, respectively. FISH was also performed, using PML and RARα probes (Oncor, Gaithersburg, MD), which detect the formation of the reciprocal RARα-PML fusion gene on del(17q) in APL cases associated with the t(15; 17), in addition to single signals relating to the normal PML and RARα loci. To further characterize cases with cryptic PML-RARα fusion genes, whole chromosome paints (WCPs; Vysis, Richmond, Surrey, UK) and biotinylated centromere probes (Oncor) for chromosomes 15 and 17 were used, in accordance with the manufacturer's instructions. Images were captured with a Zeiss Axioskop epifluorescence microscope and cooled CCD camera (Photometrics, AZ), controlled by an Apple Macintosh computer (Apple Computer Inc, Cupertino, CA). Image capture and processing software were obtained from Vysis, UK.

Immunofluorescence. The PML immunofluorescence technique as applied to crude bone marrow or peripheral blood smears using polyclonal PML antisera has been described in detail previously.5 Dual immunofluorescence studies were also undertaken using a polyclonal antiserum raised against the PML nuclear body constituent, PIC 1,24 and a monoclonal antibody (PG-M3; kindly provided by P.G. Pelicci, European Institute of Oncology, Milan, Italy) directed against the amino-terminal of PML, which recognizes both wild-type PML and the PML-RARα fusion protein.52 Bone marrow/peripheral blood smears were fixed in methanol at −20°C for 15 minutes, allowed to air dry, and preblocked with 10% fetal calf serum. Slides were then incubated simultaneously with PG-M3 and PIC 1 antibodies at a dilution of 1/5 and 1/200, respectively, in phosphate-buffered saline (PBS)-Tween 0.5% for 1 hour at room temperature. Slides were subsequently incubated with Texas-Red–coupled antimouse (Dako Ltd, High Wycombe, UK) and fluorescein-coupled antirabbit (Amersham Intl, Amersham, UK) secondary antibodies, each at 1/200 dilution, for 30 minutes at room temperature. All incubations were followed by three washes in PBS followed by a final wash comprising 0.05% Tween in PBS for 10 minutes. Preparations were examined by confocal laser scanning as previously described.5 24 

Cytogenetic and molecular characterization of APL cases lacking the t(15; 17). Cytogenetic and molecular findings in 7 patients with morphologic features of APL, but lacking the t(15; 17), are presented in Table 1. In 6 of 7 patients, RT-PCR confirmed the presence of a PML-RARα rearrangement, and in each of these cases PML-RARα transcripts were detected in the absence of RARα-PML. In the remaining patient (case no. 7), APL was associated with t(11; 17) (q23; q12-21); RT-PCR confirmed expression of both PLZF-RARα and reciprocal RARα-PLZF transcripts (Fig 1). Sequencing of PLZF-RARα and RARα-PLZF PCR products was consistent with breakpoints within the second intron of RARα and within the intron separating the exons coding for the second and third zinc fingers of PLZF, as identified in 5 of 6 previously described cases.41 53 

Fig. 1.

Detection of PLZF-RARα and RARα-PLZF transcripts by nested RT-PCR in t(11; 17)(q23; q12-21)-associated APL. PLZF-RARα and RARα-PLZF cDNA sequences are shown on the right; the positions of RARα and PLZF fusion junctions are delineated by vertical arrows.

Fig. 1.

Detection of PLZF-RARα and RARα-PLZF transcripts by nested RT-PCR in t(11; 17)(q23; q12-21)-associated APL. PLZF-RARα and RARα-PLZF cDNA sequences are shown on the right; the positions of RARα and PLZF fusion junctions are delineated by vertical arrows.

Close modal

FISH analysis of APL cases with cryptic PML-RARα rearrangements. FISH using ICRF probes, PML cos 15.5 and RARα cos 121, confirmed formation of a PML-RARα fusion in each of the 6 APL cases in which PML-RARα rearrangements were identified by RT-PCR (Table 1). In 4 patients with available diagnostic metaphase spreads, the mechanism underlying the PML-RARα rearrangement was further characterized, using ICRF cosmid probes that specifically detect the PML-RARα fusion gene, in conjunction with centromere probes and WCPs for chromosomes 15 and 17. In 3 patients, in each of whom morphologically normal chromosomes 15 and 17 were identified by conventional cytogenetics (cases no. 1 through 3, Table 1), the PML-RARα fusion gene was localized to chromosome 15q (Fig 2a and b). In each case, 15 and 17 specific paints hybridized solely to their respective chromosomes (Fig 2c and d), consistent with an interstitial insertion of RARα into PML on 15q. Furthermore, in each of these cases, using commercially available probes (Oncor), RARα was found to hybridize to two normal appearing chromosome 17s in addition to forming a fusion signal on 15q, again consistent with formation of a PML-RARα fusion on 15q and absence of the reciprocal RARα-PML fusion gene as suggested by RT-PCR analyses. In the remaining patient with evaluable metaphases, PML 15.5 and RARα 121 probes localized the PML-RARα fusion to 17q (case no. 4; Fig 3a and b). In addition to the PML-RARα fusion signal, a more centromeric RARα hybridization signal was observed. WCPs demonstrated insertion of chromosome 15 material into 17q, such that chromosome 17 appeared abnormally large on conventional cytogenetic assessment (Fig 3c and d). These results indicate that the PML-RARα fusion in this patient reflected insertion of PML with more centromeric chromosome 15-derived material into the genomic region spanned by RARα cos 121; again, this was consistent with detection of PML-RARα in the absence of RARα-PML fusion transcripts by RT-PCR.

Fig. 2.

Cryptic PML-RARα fusion resulting from interstitial insertion of RARα into PML on 15q (case no. 2; Table 1). (a) FISH analysis using ICRF PML 15.5 (green) and RARα 121 (red) cosmid probes. The RARα probe hybridized to two chromosome 17s of normal appearance, whereas 1 normal PML locus was observed on chromosome 15. The PML-RARα fusion was detected on 15q (yellow arrow). Localization of the fusion gene was confirmed by subsequent hybridization with a chromosome 15 centromere probe, shown in red in (b). (c) Chromosome 15 paint (red); chromosome 17 centromere probe (green). (d) Chromosome 17 paint (green); chromosome 15 centromere probe (red)

Fig. 2.

Cryptic PML-RARα fusion resulting from interstitial insertion of RARα into PML on 15q (case no. 2; Table 1). (a) FISH analysis using ICRF PML 15.5 (green) and RARα 121 (red) cosmid probes. The RARα probe hybridized to two chromosome 17s of normal appearance, whereas 1 normal PML locus was observed on chromosome 15. The PML-RARα fusion was detected on 15q (yellow arrow). Localization of the fusion gene was confirmed by subsequent hybridization with a chromosome 15 centromere probe, shown in red in (b). (c) Chromosome 15 paint (red); chromosome 17 centromere probe (green). (d) Chromosome 17 paint (green); chromosome 15 centromere probe (red)

Close modal
Fig. 3.

PML-RARα fusion resulting from interstitial insertion of PML with associated chromosome 15-derived material into RARα on 17q (case no. 4; Table 1). (a) FISH analysis using ICRF PML (green) and RARα (red) cosmid probes. PML-RARα fusion gene was detected on 17q (yellow arrow), adjacent to RARα hybridization signal (red arrow). Splitting of the RARα- derived signal on der(17q) was indicative of insertion of PML and adjacent sequence into the genomic region covered by RARα cosmid 121. Localization of the PML-RARα fusion gene was confirmed by subsequent hybridization with a chromosome 17 centromere probe shown in green in (b). (c) Chromosome 15 paint (red) and chromosome 17 centromere probe (green), confirming insertion of chromosome 15-derived material into 17q. (d) Chromosome 17 paint (green), chromosome 15 centromere probe (red). Chromosome 17 paint remained localized to 17; der(17q) showed a region of absent signal corresponding to the inserted region of 15q shown in (c).

Fig. 3.

PML-RARα fusion resulting from interstitial insertion of PML with associated chromosome 15-derived material into RARα on 17q (case no. 4; Table 1). (a) FISH analysis using ICRF PML (green) and RARα (red) cosmid probes. PML-RARα fusion gene was detected on 17q (yellow arrow), adjacent to RARα hybridization signal (red arrow). Splitting of the RARα- derived signal on der(17q) was indicative of insertion of PML and adjacent sequence into the genomic region covered by RARα cosmid 121. Localization of the PML-RARα fusion gene was confirmed by subsequent hybridization with a chromosome 17 centromere probe shown in green in (b). (c) Chromosome 15 paint (red) and chromosome 17 centromere probe (green), confirming insertion of chromosome 15-derived material into 17q. (d) Chromosome 17 paint (green), chromosome 15 centromere probe (red). Chromosome 17 paint remained localized to 17; der(17q) showed a region of absent signal corresponding to the inserted region of 15q shown in (c).

Close modal

Nuclear architecture in APL cases lacking the t(15; 17). Immunofluorescence studies were performed using polyclonal PML antisera. In 3 patients, cryptic PML-RARα rearrangements detected by FISH and RT-PCR were confirmed by the presence of the characteristic microparticulate nuclear staining pattern in leukemic cells (Table 1 and Fig 4a), identical to that observed in the NB4 cell-line and APL cases associated with the t(15; 17)5; whereas, in the APL case associated with the t(11; 17) leading to a PLZF-RARα rearrangement, a wild-type pattern of PML nuclear staining was observed within the leukemic blasts (Fig 4c), identical to that seen in 2 non-APL AML patients (Fig 4b) and to that previously described for HL60 and U937 cell-lines.5 

Fig. 4.

PML immunofluorescence in AML using polyclonal antisera. Phase contrast is shown in left-hand panels and corresponding PML immunofluorescence on the right. In APL cases with cryptic PML-RARα rearrangements, a microparticulate pattern of PML nuclear staining was observed as shown in (a). In non-APL cases, a wild-type pattern of PML nuclear staining was detected, as shown in AML M2 blasts in (b); similar nuclear staining was observed in t(11; 17)-associated APL (case no. 7; Table 1), shown in (c).

Fig. 4.

PML immunofluorescence in AML using polyclonal antisera. Phase contrast is shown in left-hand panels and corresponding PML immunofluorescence on the right. In APL cases with cryptic PML-RARα rearrangements, a microparticulate pattern of PML nuclear staining was observed as shown in (a). In non-APL cases, a wild-type pattern of PML nuclear staining was detected, as shown in AML M2 blasts in (b); similar nuclear staining was observed in t(11; 17)-associated APL (case no. 7; Table 1), shown in (c).

Close modal

Dual immunofluorescence studies were subsequently undertaken using a PML monoclonal antibody and polyclonal antiserum directed against PIC 1, a newly described constituent of PML nuclear bodies. In 3 non-APL AML cases, PML and PIC 1 were colocalized within discrete nuclear bodies as shown in Fig 5b; as distinct from the pattern detected in a patient with a cryptic PML-RARα rearrangement (case no. 5) in which microparticulate PML staining was observed, with only partial colocalization with PIC 1 (Fig 5a). In the patient with t(11; 17)-associated APL, PML and PIC 1 were colocalized within discrete nuclear bodies (Fig 5c), as observed in non-APL cases. In addition to discrete punctate nuclear staining, PIC 1 was also localized to the nuclear membrane, as clearly shown in Fig 5b and c.

Fig. 5.

PML and PIC 1 localization in AML. Dual immunofluorescence using PML monoclonal (left-hand panel) and PIC 1 polyclonal (center panel) antibodies. Images are fused in the right-hand panel; yellow signal denotes regions of PML/PIC 1 colocalization. (a) In APL cases associated with cryptic PML-RARα rearrangements, a microparticulate pattern of PML nuclear staining was associated with partial colocalization with PIC 1. In non-APL cases, eg, AML M4, shown in (b) and in t(11; 17)-associated APL (case no. 7; Table 1), shown in (c), PIC 1 was localized to the nuclear membrane and colocalized with PML within discrete nuclear bodies.

Fig. 5.

PML and PIC 1 localization in AML. Dual immunofluorescence using PML monoclonal (left-hand panel) and PIC 1 polyclonal (center panel) antibodies. Images are fused in the right-hand panel; yellow signal denotes regions of PML/PIC 1 colocalization. (a) In APL cases associated with cryptic PML-RARα rearrangements, a microparticulate pattern of PML nuclear staining was associated with partial colocalization with PIC 1. In non-APL cases, eg, AML M4, shown in (b) and in t(11; 17)-associated APL (case no. 7; Table 1), shown in (c), PIC 1 was localized to the nuclear membrane and colocalized with PML within discrete nuclear bodies.

Close modal

Early studies in specialist cytogenetic centers reported that the t(15; 17) could be detected in all cases of APL.42 In the light of such claims, clinicians encountering AML cases with morphologic features of APL could lose faith in the initial clinical diagnosis if subsequent cytogenetic assessment failed to provide appropriate confirmatory evidence. However, since the characterization of the PML-RARα rearrangement that underlies the t(15; 17) and identification of the rare variant translocations whereby RARα is fused to partners other than PML, it is clear that absence of the t(15; 17) does not preclude a morphologic diagnosis of APL, although it remains the diagnostic hallmark of the disease. Large multicenter studies such as the UK MRC ATRA trial afford the opportunity to determine the frequency of cryptic rearrangements and variant translocations among patients with suspected APL. In this regard, in only 87% of APL patients with molecular evidence of a PML-RARα rearrangement was the t(15; 17) detectable by conventional cytogenetics. In most cases, absence of the t(15; 17) was a reflection of failed cytogenetics; however, 2% of cases were due to cryptic PML-RARα rearrangements.5 

In the present study, we have characterized a series of 7 patients with morphologic APL found to lack the t(15; 17) on successful conventional cytogenetic assessment. In 6 patients, of whom 5 had morphologically normal chromosomes 15 and 17, the diagnosis was confirmed by the presence of a PML-RARα rearrangement; in each case, PML-RARα transcripts were detected by RT-PCR in the absence of the reciprocal derived RARα-PML species. FISH analyses in 4 such patients with evaluable metaphase spreads confirmed the presence of a nonreciprocal translocation and in each case were consistent with formation of PML-RARα as the sole fusion gene as a result of an interstitial insertion event, most commonly due to insertion of RARα into PML on 15q. This phenomenon was observed in 3 cases and has been the subject of two previous case reports in which chromosomes 15 and 17 also appeared normal by conventional cytogenetics.6 44 In the remaining patient with evaluable metaphases in the present study, the PML-RARα fusion was found to result from insertion of PML into RARα on 17q, which has not been previously described. Future characterization of the genomic breakpoints of these cases may provide insights not only into mechanisms mediating interstitial insertions but also into mechanisms underlying the more typical classical reciprocal translocation.

Demonstration of PML-RARα as the sole fusion gene formed in each of the APL cases with cryptic PML-RARα rearrangements in this study is consistent with the proposed role of its gene product as a critical mediator of leukemogenesis.2,43,51 This has recently been confirmed in a transgenic model3,4 whereby expression of PML-RARα was associated with impairment of normal myeloid differentiation accompanied by accumulation of primitive precursors and predisposition to the development of an APL-like syndrome responsive to ATRA. Although the presence of a latent period before developing the leukemia argues in favor of a requirement for additional mutational events, as has been suggested in theoretical models of tumorigenesis,54 it could also imply that high-level expression of the transgene did not occur within equivalent progenitors to those forming the targets of leukemic transformation in human APL. Although PML-RARα is clearly established as playing a key role in leukemogenesis and determining the differentiation response to ATRA, any role for RARα-PML in the pathogenesis of the disease remains to be determined. Although a single case of APL in which a RARα-PML fusion gene was apparently formed in isolation has been reported,6 the present study demonstrating a series of cases in which PML-RARα was the sole fusion gene formed argues against a significant role for RARα-PML in the pathogenesis of APL. Furthermore, this study, when considered in conjunction with previous series establishing that RARα-PML transcripts are not detectable in approximately 20% APL patients,5 shows that, at least in a proportion of cases, absence of RARα-PML transcripts reflects the occurrence of PML-RARα rearrangements due to interstitial insertion events rather than the classic reciprocal translocation.

In 1 patient in the present study, APL was associated with t(11; 17)(q23; q12-21), leading to a PLZF-RARα rearrangement. In common with the index case,53 both PLZF-RARα and RARα-PLZF transcripts could be detected by RT-PCR, consistent with the concept that, in contrast to t(15; 17)-mediated APL, both fusion partners are implicated in leukemogenesis.41,55 Whereas PLZF bears no structural similarities to PML, being characterized by an amino-terminal POZ motif, and 9 carboxy-terminal zinc finger domains implicated in its role as a transcription factor41,53,55-58; the two proteins and their respective fusion products share a number of common features. In particular, PLZF is localized to discrete nuclear bodies56,58 whose formation is dependent on the integrity of the POZ domain57 and exhibits growth-suppressor activity in transformation assays.48,59 Furthermore, in common with PML-RARα, PLZF-RARα may sequester RXR and binds retinoid response elements inhibiting transactivation.56,57,60 These processes involving disrupted growth-suppressor function coupled with deregulation of retinoid signaling pathways appear to provide a final common pathway for the pathogenesis of APL. However, in marked contrast to the disease associated with PML-RARα rearrangements, in vitro differentiation assays have shown that blasts derived from t(11; 17)(q23; q21) APL cases are resistant to retinoids.41 This may be accounted for by a number of factors, including differing repertoire and character of response between the respective fusion proteins in the presence of ligand,57,60,61 possibly compounded by the absence of ATRA-induced degradation of PLZF-RARα34 and by upregulation of RARα-PLZF leading to persistent deregulation of the cell cycle.41,55 59 

Recent studies suggest that there is at least partial colocalization of PML and PLZF within the nucleus,61,62 raising the possibilities that their growth-suppressor activities might be interrelated and that fusion proteins associated with variant APL translocations could promote leukemogenesis by disruption of PML nuclear bodies. Therefore, in the context of such a model, one might expect ATRA-resistant t(11; 17) cases to maintain a disrupted pattern of PML nuclear bodies, reminiscent of that reported in retinoid-resistant PML-RARα–mediated cases.19,35 However, the present study refutes such a hypothesis; disruption of PML nuclear bodies was only observed in APL cases associated with cryptic PML-RARα rearrangements in which PML nuclear staining patterns were identical to t(15; 17)-positive cases, whereas a wild-type pattern of PML nuclear staining was detected in blasts derived from t(11; 17)(q23; q21)-associated APL, reminiscent of that observed in other subtypes of AML. Furthermore, using dual immunofluorescence techniques, we were able to show that PIC 1 (GMP 1,63 SUMO 164), a novel-ubiquitin homology domain protein identified as an interaction partner of PML in a yeast-two hybrid screen,24 interacts with PML in vivo in AML blasts as well as in nonhemopoietic cell lines, as previously described.24 In t(11; 17)-associated APL and in non-APL cases, in addition to perfect colocalization with PML within discrete nuclear bodies, a perinuclear pattern of PIC 1 staining was observed consistent with its reported interaction with RanGap1, targeting it to the nuclear pore complex.63,64 In contrast, in APL cases associated with cryptic PML-RARα rearrangements, only partial colocalization of PML and PIC 1 was observed, confirming results previously obtained with the APL cell-line NB4,24 reflecting disruption of PML nuclear bodies in the presence of the PML-RARα fusion protein. Recent work has established that the second heptad repeat of the coiled-coil domain of PML-RARα is critical for nuclear body disruption, which does not appear to be necessary either for the block in differentiation or its reversal by retinoids; both of these effects are dependent on the integrity of the first heptad repeat of the coiled-coil region.29 Whether this perturbation of nuclear architecture is involved in the process of leukemic transformation is still undetermined. It remains a possibility that disruption of the nuclear bodies is merely a secondary phenomenon reflecting an interaction between the PML-RARα fusion protein and wild-type PML and is of no importance to leukemogenesis. Nevertheless, the present study underlines the fact that disruption of PML nuclear bodies provides a valuable marker for the PML-RARα fusion protein in cases lacking the t(15; 17) and confirms PML immunofluorescence as a suitable technique for rapid identification of the subgroup of APL patients likely to benefit from retinoids.5 65 In conclusion, although deregulation of the putative growth-suppressor PML and delocalization of other nuclear body constituents have been advocated to play a key role in the development of t(15; 17)-associated APL, the present study shows that disruption of PML nuclear bodies per se is not a prerequisite for the pathogenesis of APL.

A wild-type PML nuclear localization pattern has also recently been reported in an APL case with the variant translocation t(11;17)(q13;q21) leading to a NUMA/RARα rearrangement.47 PIC 1 has been designated UBL 1 by the Gene Nomenclature Committee.

The authors are grateful to all the clinicians who entered patients into the MRC ATRA trial and forwarded material for molecular and cytogenetic analyses, particularly Dr Steve Kelsey. We thank Steve Chatters and Joanne Rogers in the Cytogenetics Laboratory at University College Hospital, London, and the cytogeneticists involved in karyotyping these patients who provided material for FISH analyses, particularly Debra Lillington, Michael Neat, and David Stevenson. We are grateful to Peter Jordan for assistance with immunofluorescence studies, to the photographic department at ICRF, Lincoln's Inn Fields, and to Iain Goldsmith and the oligonucleotide synthesis service at ICRF, Clare Hall. We also thank Hans Nicolai, Dr Melissa Brown, Dr Aurélie Catteau, and Dr Chun-Fang Xu for helpful discussions and assistance with sequence analyses.

D.G. was supported by an MRC clinical training fellowship. E.S. and K.H. were supported by EEC grants BIOMED-CT92-0755 and Biotech BI02-CT-930450. E.D. was supported by an EC TMR fellowship. D.G. and K.H. are currently supported by ICRF. S.L. and DNA/RNA banking facilities at University College Hospital, London are currently supported by the Kay Kendall Leukaemia Fund.

Address reprint requests to Dr David Grimwade, Cancer Genetics Laboratory, UMDS, 8th Floor, Guy's Tower, Guy's Hospital, London SE1 9RT, UK.

1
Rowley
JD
Golomb
HM
Dougherty
C
15/17 translocation, a consistent chromosomal change in acute promyelocytic leukaemia.
Lancet
1
1977
549
2
Grimwade
D
Solomon
E
Characterisation of the PML/RARα rearrangement associated with t(15; 17) acute promyelocytic leukaemia.
Curr Topics Microbiol Immunol
220
1997
81
3
Brown
D
Kogan
S
Lagasse
E
Weissman
I
Alcalay
M
Pelicci
PG
Atwater
S
Bishop
JM
A PMLRARα transgene initiates murine acute promyelocytic leukemia.
Proc Natl Acad Sci USA
94
1997
2551
4
Grisolano
JL
Wesselschmidt
RL
Pelicci
PG
Ley
TJ
Altered myeloid development and acute leukemia in transgenic mice expressing PML-RARα under control of cathepsin G regulatory sequences.
Blood
89
1997
376
5
Grimwade
D
Howe
K
Langabeer
S
Davies
L
Oliver
F
Walker
H
Swirsky
D
Wheatley
K
Goldstone
A
Burnett
A
Solomon
E
Establishing the presence of the t(15; 17) in suspected acute promyelocytic leukaemia: Cytogenetic, molecular and PML immunofluorescence assessment of patients entered into the M.R.C. ATRA trial.
Br J Haematol
94
1996
557
6
Lafage-Pochitaloff
M
Alcalay
M
Brunel
V
Longo
L
Sainty
D
Simonetti
J
Birg
F
Pelicci
PG
Acute promyelocytic leukemia cases with nonreciprocal PML/RARα or RARα /PML fusion genes.
Blood
85
1995
1169
7
Grignani
F
Ferrucci
PF
Testa
U
Talamo
G
Fagioli
M
Alcalay
M
Mencarelli
A
Peschle
C
Nicoletti
I
Pelicci
PG
The acute promyelocytic leukemia-specific PML-RARα fusion protein inhibits differentiation and promotes survival of myeloid precursor cells.
Cell
74
1993
423
8
Miller
WH
Kakizuka
A
Frankel
SR
Warrell
RP
DeBlasio
A
Levine
K
Evans
R
Dmitrovsky
E
Reverse transcription polymerase chain reaction for the rearranged retinoic acid receptor α clarifies diagnosis and detects minimal residual disease in acute promyelocytic leukemia.
Proc Natl Acad Sci USA
89
1992
2694
9
Stunnenberg
HG
Mechanisms of transactivation by retinoic acid receptors.
Bioessays
15
1993
309
10
Kliewer
SA
Umesono
K
Mangelsdorf
DJ
Evans
RM
Retinoid X receptor interacts with nuclear receptors in retinoic acid, thyroid hormone and vitamin D3 signalling.
Nature
355
1992
446
11
Zhang
X-K
Hoffmann
B
Tran
PB-V
Graupner
G
Pfahl
M
Retinoid X receptor is an auxiliary protein for thyroid hormone and retinoic acid receptors.
Nature
355
1992
441
12
Tsai
S
Collins
SJ
A dominant negative retinoic acid receptor blocks neutrophil differentiation at the promyelocyte stage.
Proc Natl Acad Sci USA
90
1993
7153
13
Onodera
M
Kunisada
T
Nishikawa
S
Sakiyama
Y
Matsumoto
S
Nishikawa
S-I
Overexpression of retinoic acid receptor α suppresses myeloid cell differentiation at the promyelocyte stage.
Oncogene
11
1995
1291
14
Kakizuka
A
Miller
WH
Umesono
K
Warrell
RP
Frankel
SR
Murty
VVVS
Dmitrovsky
E
Evans
RM
Chromosomal translocation t(15; 17) in human acute promyelocytic leukemia fuses RARα with a novel putative transcription factor, PML.
Cell
66
1991
663
15
Goddard
AD
Borrow
J
Freemont
PS
Solomon
E
Characterization of a zinc finger gene disrupted by the t(15; 17) in acute promyelocytic leukemia.
Science
254
1991
1371
16
Saurin
AJ
Borden
KLB
Boddy
MN
Freemont
PS
Does this have a familiar RING?
Trends Biochem Sci
21
1996
208
17
Daniel
MT
Koken
M
Romagné
O
Barbey
S
Bazarbachi
A
Stadler
M
Guillemin
MC
Degos
L
Chomienne
C
de Thé
H
PML protein expression in hematopoietic and acute promyelocytic leukemia cells.
Blood
82
1993
1858
18
Weis
K
Rambaud
S
Lavau
C
Jansen
J
Carvalho
T
Carmo-Fonseca
M
Lamond
A
Dejean
A
Retinoic acid regulates aberrant nuclear localization of PML-RARα in acute promyelocytic leukaemia cells.
Cell
76
1994
345
19
Dyck
JA
Maul
GG
Miller
WH
Chen
JD
Kakizuka
A
Evans
RM
A novel macromolecular structure is a target of the promyelocyte-retinoic acid receptor oncoprotein.
Cell
76
1994
333
20
Koken
MHM
Puvion-Dutilleul
F
Guillemin
MC
Viron
A
Linares-Cruz
G
Stuurman
N
de Jong
L
Szostecki
C
Calvo
F
Chomienne
C
Degos
L
Puvion
E
de Thé
H
The t(15; 17) translocation alters a nuclear body in a retinoic acid-reversible fashion.
EMBO J
13
1994
1073
21
Korioth
F
Gieffers
C
Maul
GG
Frey
J
Molecular characterization of NDP52, a novel protein of the nuclear domain 10, which is redistributed upon virus infection and interferon treatment.
J Cell Biol
130
1995
1
22
Szostecki
C
Guldner
HH
Netter
HJ
Will
H
Isolation and characterization of cDNA encoding a human nuclear antigen predominantly recognized by autoantibodies from patients with primary biliary cirrhosis.
J Immunol
145
1990
4338
23
Bloch
DB
de la Monte
SM
Guigaouri
P
Filippov
A
Bloch
KD
Identification and characterization of a leukocyte-specific component of the nuclear body.
J Biol Chem
271
1996
29198
24
Boddy
MN
Howe
K
Etkin
LD
Solomon
E
Freemont
PS
PIC 1, a novel ubiquitin-like protein which interacts with the PML component of a multiprotein complex that is disrupted in acute promyelocytic leukaemia.
Oncogene
13
1996
971
25
Mu
Z-M
Chin
K-V
Liu
J-H
Lozano
G
Chang
K-S
PML, a growth suppressor disrupted in acute promyelocytic leukemia.
Mol Cell Biol
14
1994
6858
26
Koken
MHM
Linares-Cruz
G
Quignon
F
Viron
A
Chelbi-Alix
MK
Sobczak-Thépot
J
Juhlin
L
Degos
L
Calvo
F
de Thé
H
The PML growth-suppressor has an altered expression in human oncogenesis.
Oncogene
10
1995
1315
27
Liu
J-H
Mu
Z-M
Chang
K-S
PML suppresses oncogenic transformation of NIH/3T3 cells by activated neu.
J Exp Med
181
1995
1965
28
Chang
K-S
Fan
Y-H
Andreeff
M
Liu
J
Mu
Z-M
The PML gene encodes a phosphoprotein associated with the nuclear matrix.
Blood
85
1995
3646
29
Grignani
F
Testa
U
Rogaia
D
Ferrucci
PF
Samoggia
P
Pinto
A
Aldinucci
D
Gelmetti
V
Fagioli
M
Alcalay
M
Seeler
J
Grignani
F
Nicoletti
I
Peschle
C
Pelicci
PG
Effects on differentiation by the promyelocytic leukemia PML/RARα protein depend on the fusion of the PML protein dimerization and RARα DNA binding domains.
EMBO J
15
1996
4949
30
de Thé
H
Lavau
C
Marchio
A
Chomienne
C
Degos
L
Dejean
A
The PML-RARα fusion mRNA generated by the t(15; 17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR.
Cell
66
1991
675
31
Pandolfi
PP
Grignani
F
Alcalay
M
Mencarelli
A
Biondi
A
Lo
Coco F
Grignani
F
Pelicci
PG
Structure and origin of the acute promyelocytic leukemia myl/RARα cDNA and characterization of its retinoid-binding and transactivation properties.
Oncogene
6
1991
1285
32
Kastner
P
Perez
A
Lutz
Y
Rochette-Egly
C
Gaub
M-P
Durand
B
Lanotte
M
Berger
R
Chambon
P
Structure, localisation and transcriptional properties of two classes of retinoic acid receptor α fusion proteins in acute promyelocytic leukemia (APL): Structural similarities with a new family of oncoproteins.
EMBO J
11
1992
629
33
Raelson
JV
Nervi
C
Rosenauer
A
Benedetti
L
Monczak
Y
Pearson
M
Pelicci
PG
Miller
WH
The PML/RARα oncoprotein is a direct molecular target of retinoic acid in acute promyelocytic leukemia cells.
Blood
88
1996
2826
34
Yoshida
H
Kitamura
K
Tanaka
K
Omura
S
Miyazaki
T
Hachiya
T
Ohno
R
Naoe
T
Accelerated degradation of PML-Retinoic acid receptor α (PML-RARA) oncoprotein by all-trans-retinoic acid in acute promyelocytic leukemia: Possible role of the proteosome pathway.
Cancer Res
56
1996
2945
35
Rosenauer
A
Raelson
JV
Nervi
C
Eydoux
P
DeBlasio
A
Miller
WH
Alterations in expression, binding to ligand and DNA, and transcriptional activity of rearranged and wild-type retinoid receptors in retinoid-resistant acute promyelocytic leukemia cell-lines.
Blood
88
1996
2671
36
Warrell
RP
Frankel
SR
Miller
WH
Scheinberg
DA
Itri
LM
Hittelman
WN
Vyas
R
Andreeff
M
Tafuri
A
Jakubowski
A
Gabrilove
J
Gordon
MS
Dmitrovsky
E
Differentiation therapy for acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid).
N Engl J Med
324
1991
1385
37
Chen
Z-X
Xue
Y-Q
Zhang
R
Tao
R
Xia
X
Li
C
Wang
W
Zu
W
Yao
X
Ling
B
A clinical and experimental study on all-trans-retinoic acid-treated acute promyelocytic leukemia patients.
Blood
78
1991
1413
38
Castaigne
S
Chomienne
C
Daniel
MT
Ballerini
P
Berger
R
Fenaux
P
Degos
L
All-trans retinoic acid as a differentiation therapy for acute promyelocytic leukemia. I. Clinical results.
Blood
76
1990
1704
39
Fenaux P, Chastang C, Castaigne S, Archimbaud E, Sanz M, Link H, Guerci A, Fegueux N, Zittoun R, Stoppa AM, Travade P, Lamy T, Maloisel F, Sadoun A, San Miguel J, Veil A, Rayon C, Conde E, Fey M, Bordessoule D, Ganser A, Bowen D, Dreyfus F, Huguet F, Tilly H, Guy H, Auzanneau G, Chomienne C, Degos L: Treatment of newly diagnosed acute promyelocytic leukemia (APL) with all-transretinoic acid (ATRA) followed by intensive chemotherapy (CT). Updated results of the European group. Blood 84:379a, 1994 (abstr, suppl 1)
40
Tallman MS, Andersen J, Schiffer CA, Appelbaum FR, Feusner JE, Woods WG, Ogden A, Weinstein H, Shepherd L, Rowe JM, Wiernik PH: Phase III randomized study of all-trans retinoic acid (ATRA) vs daunorubicin (D) and cytosine arabinoside (A) as induction therapy and ATRA vs observation as maintenance therapy for patients with previously untreated acute promyelocytic leukemia (APL). Blood 86:125a, 1995 (abstr, suppl 1)
41
Licht
JD
Chomienne
C
Goy
A
Chen
A
Scott
AA
Head
DR
Michaux
JL
Wu
Y
DeBlasio
A
Miller
WH
Zelenetz
AD
Willman
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
42
Larson
RA
Kondo
K
Vardiman
JW
Butler
AE
Golomb
HM
Rowley
JD
Evidence for a 15; 17 translocation in every patient with acute promyelocytic leukemia.
Am J Med
76
1984
827
43
Borrow
J
Shipley
J
Howe
K
Kiely
F
Goddard
A
Sheer
D
Srivastava
A
Antony
AC
Fioretos
T
Mitelman
F
Solomon
E
Molecular analysis of simple variant translocations in acute promyelocytic leukaemia.
Genes Chromosom Cancer
9
1994
234
44
Hiorns
LR
Min
T
Swansbury
GJ
Zelent
A
Dyer
MJS
Catovsky
D
Interstitial insertion of retinoic acid receptor-α gene in acute promyelocytic leukemia with normal chromosomes 15 and 17.
Blood
83
1994
2946
45
Chen
S-J
Zelent
A
Tong
J-H
Yu
H-Q
Wang
Z-Y
Derré
J
Berger
R
Waxman
S
Chen
Z
Rearrangements of the retinoic acid receptor alpha and promyelocytic zinc finger genes resulting from t(11; 17)(q23; q21) in a patient with acute promyelocytic leukaemia.
J Clin Invest
91
1993
2260
46
Redner
RL
Rush
EA
Faas
S
Rudert
WA
Corey
SJ
The t(5; 17) variant of acute promyelocytic leukemia expresses a nucleophosmin-retinoic acid receptor fusion.
Blood
87
1996
882
47
Wells
RA
Catzavelos
C
Kamel-Reid
S
Fusion of retinoic acid receptor α to NUMA, the nuclear mitolic apparatus protein, by a variant translocation in acute promyelocytic leukaemia.
Nat Genet
17
1997
109
48
Pandolfi
PP
PML, PLZF and NPM genes in the molecular pathogenesis of acute promyelocytic leukemia.
Haematologica
81
1996
472
49
Webber
LM
Garson
OM
Fluorodeoxyuridine synchronization of bone marrow cultures.
Cancer Genet Cytogenet
8
1982
123
50
Grimwade
D
Howe
K
Langabeer
S
Burnett
A
Goldstone
A
Solomon
E
Minimal residual disease detection in acute promyelocytic leukemia by reverse-transcriptase PCR: Evaluation of PML-RARα and RARα-PML assessment in patients who ultimately relapse.
Leukemia
10
1996
61
51
Borrow
J
Goddard
AD
Gibbons
B
Katz
F
Swirsky
D
Fioretos
T
Dube
I
Winfield
DA
Kingston
J
Hagemeijer
A
Rees
JKH
Lister
TA
Solomon
E
Diagnosis of acute promyelocytic leukaemia by RT-PCR: Detection of PML-RARA and RARA-PML fusion transcripts.
Br J Haematol
82
1992
529
52
Flenghi
L
Fagioli
M
Tomassoni
L
Pileri
S
Gambacorta
M
Pacini
R
Grignani
F
Casini
T
Ferrucci
PF
Martelli
MF
Pelicci
PG
Falini
B
Characterization of a new monoclonal antibody (PG-M3) directed against the aminoterminal portion of the PML gene product: Immunocytochemical evidence for high expression of PML proteins on activated macrophages, endothelial cells, and epithelia.
Blood
85
1995
1871
53
Chen
Z
Brand
NJ
Chen
A
Chen
S-J
Tong
J-H
Wang
Z-Y
Waxman
S
Zelent
A
Fusion between a novel Krüppel-like zinc finger gene and the retinoic acid receptor-α locus due to a variant t(11; 17) translocation associated with acute promyelocytic leukaemia.
EMBO J
12
1993
1161
54
Vickers M: Estimation of the number of mutations necessary to cause chronic myeloid and acute promyelocytic leukaemias from epidemiologic data. Br J Haematol 93:60, 1996 (abstr, suppl 1)
55
Sitterlin
D
Tiollais
P
Transy
C
The RARα-PLZF chimera associated with acute promyelocytic leukemia has retained a sequence-specific DNA-binding domain.
Oncogene
14
1997
1067
56
Licht
JD
Shaknovich
R
English
MA
Melnick
A
Li
J-Y
Reddy
JC
Dong
S
Chen
S-J
Zelent
A
Waxman
S
Reduced and altered DNA-binding and transcriptional properties of the PLZF-retinoic acid receptor-α chimera generated in t(11; 17)-associated acute promyelocytic leukemia.
Oncogene
12
1996
323
57
Dong
S
Zhu
J
Reid
A
Strutt
P
Guidez
F
Zhong
H-J
Wang
Z-Y
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-α fusion protein.
Proc Natl Acad Sci USA
93
1996
3624
58
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
59
Yeyati PL, Shaknovich R, Zelent A, Li J, Waxman S, Licht JD: Cyclin A is a candidate target gene for the promyelocytic leukemia zinc finger protein. Blood 88:291a,1996 (abstr, suppl 1)
60
Chen
Z
Guidez
F
Rousselot
P
Agadir
A
Chen
S-J
Wang
Z-Y
Degos
L
Zelent
A
Waxman
S
Chomienne
C
PLZF-RARα fusion proteins generated from the variant t(11; 17)(q23; q21) translocation in acute promyelocytic leukaemia inhibit ligand-dependent transactivation of wild-type retinoic acid receptors.
Proc Natl Acad Sci USA
91
1994
1178
61
Ruthardt M, Testa U, Nervi C, Riganelli D, Ferrucci PF, Grignani F, Alcalay M, Puccetti E, Grignani F, Peschle C, Hoelzer D, Pelicci PG: Genetic determination of retinoic acid response in acute promyelocytic leukemia. Blood 88:551a, 1996 (abstr, suppl 1)
62
Koken M, Reid A, Quignon F, Dong S, Chen Z, Strutt P, Licht J, Waxman S, de Thé H, Zelent A: Products of the PML and PLZF genes translocated with the RARα locus in acute promyelocytic leukemia co-localize in the nucleus and interact with each other in vivo. Blood 88:553a, 1996 (abstr, suppl 1)
63
Matunis
MJ
Coutavas
E
Blobel
G
A novel ubiquitin-like modification modulates the partitioning of the Ran-GTPase-activating protein RanGAP1 between the cytosol and the nuclear pore complex.
J Cell Biol
135
1996
1457
64
Mahajan
R
Delphin
C
Guan
T
Gerace
L
Melchior
F
A small ubiquitin-related polypeptide involved in targeting RanGAP1 to nuclear pore complex protein RanBP2.
Cell
88
1997
97
65
Dyck
J
Warrell
RP
Evans
RM
Miller
WH
Rapid diagnosis of acute promyelocytic leukemia by immunohistochemical localization of PML/RAR-α protein.
Blood
86
1995
862
Sign in via your Institution