IN THE LATE 1980s and early 1990s, the elucidation of the molecular basis of acute promyelocytic leukemia (APL) emerged as a paradigm for the connection between the bench and bedside. At that time, it became apparent that APL was, among the forms of acute myeloid leukemia, uniquely sensitive to all-transretinoic acid (ATRA)1,2 and clinical trials indicated that ATRA induced complete remissions by differentiation and eventual elimination of the malignant clone (reviewed previously3-8). In 1991, it was discovered that the consistent chromosomal translocation of APL, t(15:17),9fused the retinoic acid receptor α (RARα) gene to the promyelocytic leukemia (PML) gene on chromosome 15, yielding the fusion protein PML-RARα.10-15 These data suggested that disruption of RARα function was the major cause of APL. According to this line of reasoning, retinoic acid in pharmacological doses could then overcome this pathology, leading to in vivo differentiation and clinical remission. Although this hypothesis is essentially correct, 7 years of intense investigation of the APL model have begun to uncover a more complicated picture.

APL is now associated with four different gene rearrangements, fusing RARα to the PML, promyelocytic leukemia zinc finger (PLZF), nucleophosmin (NPM), or nuclear matrix associated (NuMA) genes (Fig 1), leading to the formation of reciprocal fusion proteins (N-RARα and RARα-N). This again highlights the importance of retinoid metabolism, but also suggests that partner genes with RARα could also play important roles. In this review, we will deconstruct the APL problem by evaluating the role of RARα in normal and neoplastic myeloid development. We will examine each of the genes fused to the RARα in APL, searching for similarities and differences among the four partner proteins that may explain the distinct clinical outcome some patients with variant forms of APL. Finally, we will reconstruct the disease of APL and examine the leukemogenic functions of the RARα fusion proteins in cell culture models, animal models, and patients. We will also examine how the recent explosion of knowledge in APL has led to the development of new therapeutic agents such as arsenic trioxide16 17 and sodium butyrate.

Fig. 1.

The four chromosomal translocations associated with APL result in fusion proteins in which the B through F domains of RAR, including the DNA binding and ligand binding domains of protein, are linked C-terminal to four different nuclear proteins containing self-association domains. The t(11;17) APL syndrome linking PLZF and RAR is unique among these forms of APL in its resistance to differentiation therapy with ATRA or conventional chemotherapy.

Fig. 1.

The four chromosomal translocations associated with APL result in fusion proteins in which the B through F domains of RAR, including the DNA binding and ligand binding domains of protein, are linked C-terminal to four different nuclear proteins containing self-association domains. The t(11;17) APL syndrome linking PLZF and RAR is unique among these forms of APL in its resistance to differentiation therapy with ATRA or conventional chemotherapy.

Close modal
Fig. 2.

Functional domains of the RAR protein.

Fig. 2.

Functional domains of the RAR protein.

Close modal

Retinoids may be key for myeloid differentiation. Vitamin A-deficient mice and humans were noted to have defects in hematopoiesis18,19 and retinoids can preferentially stimulate granulopoiesis.20,21 In the early 1980s, it was noted that retinoic acid (ATRA) could induce differentiation of myeloid cell lines such as HL6022 and of primary cells from patients with APL.23 The cloning of the RARs and other members of the nuclear receptor superfamily24-26 allowed for further detailed studies into the mechanism of action of ATRA. Among the genes encoding RARs (reviewed previously24-26), RARα is identified with myeloid development.27-29 

RARα structure parallels that of other nuclear receptors and is divided into 6 evolutionarily conserved domains (A through F; Fig 2). The most highly conserved domain among nuclear receptors and retinoid receptors is the C domain, which contains two C2C2 zinc finger motifs (reviewed in Chambon30). Through this domain, RARα binds to retinoic acid response elements (RARE) located in the promoters of many genes, including those of RARα,31,32 RARβ,33,34and RARγ.35 RAREs consist of a direct repeat (A/G)G(G/T)TCA separated by 2 or 5 nucleotides. RARα binds as a heterodimer to this site along with the related retinoid X receptor protein (RXR).36-38 Heterodimerization is mediated both by the DNA binding and ligand binding domain of RARα.39,40RARα is a ligand-dependent transcription factor stimulated by ATRA, whereas its partner, RXR, responds to ATRA or 9-cis retinoic acid.41 RARα and other nuclear receptors contain two domains, AF-1 (A/B domains) and AF2 (E domain), which can cooperate to activate transcription.42 AF-1, contained within the N-terminal A/B domain, is a ligand-independent transcriptional activation domain that works in a promoter context-dependent manner.42,43 Through alternative promoter usage, the RARα protein can have two different A domains (A1 or A2). The C-terminal E domain of RARα contains the AF2 ligand-dependent transcriptional activation domain as well as a dimerization interface for RXR.39,40 43 

RARs modulate transcription through interaction with cofactors. The AF-2 domain of the protein associates with corepressor molecules in the absence of ligand. These corepressors, N-CoR and SMRT,44,45were recently shown to be part of a multiprotein repressor complex also containing the Sin3A corepressor and histone deacetylases (Pazin and Kadonaga46 and references therein). This suggests that RARs may silence certain promoters by alterations in chromatin configuration. Structural studies of RXRα and RARγ indicate that, in the presence of ligand, the AF2 changes its conformation, making new residues available30,47-49 to bind to coactivator proteins. Nuclear receptor coactivators include TIF150,51 related to the PML protein associated with t(15;17)-associated APL (see below), Trip1/sug1,52 Tif2,53 ACTR,54Src-1,55 TAFII135,56 and CBP.57-59 The functions of these coactivators are beginning to be elucidated. TIF150 interacts with the TATA binding protein (TBP), TBP-associated factors (TAFs),60 and the basal factor TFIIE.61 Murine and yeast Trip1/sug1 have DNA helicase activity,62 which could unwind DNA, whereas the CBP and ACTR cofactors have histone acetylase activity and associate with P/CAF, another histone acetylase.54,63-66 It is believed that histone acetylation leads to alterations in the conformation of chromatin and stimulation of gene transcription.67 Hence, the ligand-activated RARα can best be imagined as a multiprotein complex bound to DNA in association with RXR and a number of coactivator proteins. The ligand bound complex might then stimulate transcription though interaction with basal factors, alteration of chromatin, and unwinding of DNA.

The use of synthetic ligands specific for RARα and RXR indicate that RAR/RXR complexes that stimulate gene transcription are responsible for the pro-differentiation effect of ATRA,68-71 whereas RXR/RXR complexes cannot induce differentiation of APL cells. RARs can also repress transcription through cross-talk with other transcriptional activators, including the AP1 family of activator proteins,72 probably due to competition for limiting coactivators such as CBP.73-75 However, the most important action of RARα in myeloid differentiation is its ability to activate transcription through RAREs, because artificial ligands that inhibit AP1 activity but fail to stimulate RARE-mediated transcription fail to induce myeloid differentiation.68 76 Which key genes are stimulated to affect myeloid differentiation remain to be determined.

The importance of RARα in myeloid differentiation was underscored when Collins et al77-79 developed a HL60 cell line resistant to differentiation by ATRA. This cell line harbored a dominant negative mutant RARα with a truncation within the C-terminal AF-2 domain. Differentiation of these cells under the influence of ATRA was restored by infection with a retrovirus expressing wild-type RARα, RARβ, or RARγ.77 80 Furthermore, RXRα expression in the resistant cells restored myeloid differentiation, suggesting that the mutant receptor may have acted in a dominant negative mode by heterodimerizing with wild-type RXR and forming an inactive transcriptional complex. Overexpression of RXR overcame this block, perhaps by recruiting other isoforms of RAR to mediate the transcriptional response required for differentiation.

RARα may help program normal hematopoietic development. Erythroid induction of multipotent FDCP mixA4 cells by erythropoietin was correlated with complete downregulation of RARα expression, whereas myeloid differentiation induced by granulocyte colony-stimulating factor (G-CSF) was correlated with upregulation of RARα, particularly the RARα2 isoform.81 Introduction of an RARα mutant, with a deletion in the ligand-binding domain, into a multipotential hematopoietic cell line resulted in a switch in cell fate from the granulocyte/monocyte to the mast cell lineage.82Granulocyte-macrophage colony-stimulating factor (GM-CSF)–mediated myeloid differentiation of these cells was blocked at the promyelocyte stage, an effect that could be overcome by high doses of ATRA.83 Although truncation of the RARα within the ligand binding domain has a profound effect on myeloid differentiation, this type of mutation was not identified in a series of 118 specimens of human cancer, including a number of fresh APL specimens.84In leukemia, the RARα gene is only disrupted by the formation of chromsomal translocations yielding fusion genes (see below). The notion that the dominant negative RARα functions by sequestration of RXR into inactive complexes was supported by the finding that overexpression of wild-type RARα in murine bone marrow cultures85 led to the accumulation of promyelocytic colonies. Upon addition of ATRA, the RARα-expressing marrow colonies consisted mainly of more differentiated granulocytes. Hence, overexpression of wild-type RARα, C-terminal truncated forms of RARα and fusion proteins consisting of partners fused to the N-terminus of RARα (eg, PML-RARα; see below) can all lead to the blockade of myeloid differentiation at the promyelocyte stage when cells are grown at physiological levels (∼10−8mol/L) of ATRA. Only pharmacological levels of ATRA (10−7 to 10−6 mol/L) can overcome this block. How might this blockade occur at the molecular level? The fact that wild-type RARα as well as mutant forms of RARα can cause the block suggests a squelching mechanism.86 At low ATRA concentrations, coactivators may bind loosely to nuclear receptor/DNA complexes and be easily sequestered by high levels of free normal or aberrant receptor in the nucleoplasm. Only at pharmacological ATRA concentrations would the cofactors be drawn to target genes along with RARα and the basal transcriptional machinery. In support of this notion, in vivo footprinting of the RARβ promoter shows occupancy of the RARE only under pharmacological ATRA concentrations.87It might be predicted that forced expression of RXR and/or RARα coactivators would rescue the block by dominant negative RARα and allow differentiation to proceed at physiological ATRA concentrations.

ATRA treatment of myeloid precursor cells and other cells drives the expression of multiple classes of genes (Table 1) expressed either immediately after ATRA treatment or after some delay. ATRA-induced changes in myeloid gene expression are accompanied by inhibition of cell growth and induction of terminal differentiation and production of a mature cell ready to fight infection. The retinoic acid syndrome encountered during treatment of APL with ATRA, characterized by an increase in leukocyte count, fever, and pulmonary infiltrates, may be due to the increased adhesive characteristics of the differentiating granulocytes and secretion of cytokines.88 ATRA also downregulates the expression of procoagulants found in the undifferentiated promyleocyte, explaining the ability of differentiation therapy to treat APL without inducing hemostatic disorders (reviewed in Barbui et al89). The initial waves of leukocytes found in APL patients are derived from the malignant clone90 and function normally in vitro to kill pathogens,91 despite some abnormalities of secondary granules involved in immune function.92 93 

Many of the target genes of RARα that induce rapidly after ATRA are themselves transcription factors.38 There are RAREs in the promoters of the RARs (see above) that may help explain the ability of ATRA to induce differentiation in APL. ATRA treatment of fresh APL cells upregulates the mRNA for RARα, correlating with the presence of a RARE in the second promoter of the RARα gene.32,94Therefore, one way that ATRA may induce myeloid differentiation may be to upregulate the RAR/RXR complexes, overcoming the dominant negative PML-RARα protein. This hypothesis is supported by recent data that indicate that PML-RARα is selectively degraded in APL cells by treatment with ATRA,95,96 further shifting the balance towards the wild-type receptor. Other candidates for directly regulation by the retinoid receptors during myeloid development include members of the hox family of homeobox-containing transcription factors (reviewed previously97,98). Hox genes are expressed in myeloid cell lines in a coordinated, dynamic manner.99Enforced expression of the hox genes100-103 or disruption of their expression104 as in leukemia-associated translocations105,106 is associated with altered myeloid growth and differentiation.107 RAREs were identified in the promoters and enhancers of hoxb1108-110,hoxa1,111,112 and hoxd4.113Furthermore, specific RARs differentially regulate the homeobox genes. In embryonic carcinoma cells null for RARα, ATRA fails to induce thehoxb1,38 114 whereas RARγ null cells fail to express hoxa1. This information suggests that in APL the disruption of RARα function may alter expression of a subset of ATRA-inducible genes critical for myeloid differentiation.

Recent data indicate an interplay between retinoic acid and interferon (IFN)-mediated signaling. ATRA can rapidly induce transcription of the IFN regulatory factor-1 (IRF-1) gene. IRF-1 expression is associated with the expression of IFN and IFN-inducible genes,115cessation of cellular growth, and induction of apoptosis. Thus, IFN may potentially mediate some of the antiproliferative effects of ATRA.116,117 ATRA induction of the IRF-1 promoter is mediated by a GAS (γ-IFN activation sequence) rather than an RARE,118 signifying a role for retinoic acid in the STAT (signal transducer and activator of transcription) pathway for IFN signaling.119 ATRA rapidly induces the expression of STAT1α at the mRNA level and increases tyrosine phosphorylation of STAT1α, together leading to a large increase in DNA binding activity of the STAT1α complex to an IFN-responsive element (IRE).120 RARα and STAT1α synergized to stimulate transcription from an IRE-containing reporter plasmid, whereas PML-RARα did not, suggesting that cross-talk between the two signaling pathways may be aberrant in APL and play a role in disease pathogenesis.

Recently C/EBP-ε, a newly identified basic-zipper transcription factor that recognizes CCAAT DNA sequences, was found to be rapidly upregulated during ATRA-mediated differentiation. C/EBP-ε is the only member of this family of transcription factors expressed in the APL cell line NB4,121 suggesting that the gene may play a role in the promyelocyte stage of differentiation.122-125 HL60 cells engineered to express PML-RARα downregulate C/EBP-ε expression in the absence of retinoic acid; the C/EBP-ε gene is then upregulated when pharmacological doses of ATRA are added to the cell.126 Hence, C/EBP-ε may be a model target gene of the PML-RARα fusion protein that is inhibited in expression at ambient physiological concentrations of ATRA and stimulated in expression when cells are treated with ATRA.

ATRA is known to alter cell cycle kinetics, because it induces the differentiation of APL and other myeloid cells.127,128 ATRA treatment is associated with G1 arrest and the accumulation of hypophosphorylated forms of the retinoblastoma protein.129ATRA induces the expression of the p21WAF1/Cip1cyclin-dependent kinase inhibitor in myeloid cell lines.130,131 ATRA-mediated p21 induction may also depend on the PML protein disrupted in t(15;17) APL.132 RARα in combination with RXR binds to an imperfect RARE within the human p21 promoter and RARα can activate the p21 promoter in a ligand-dependent manner. Therefore, p21 meets criteria for a bona fide RARα target gene whose expression could decrease leukemic cell proliferation. Potentially relevant to the treatment of APL, the cytosolic retinoic acid binding protein II (CRABPII) promoter contains an RARE and can be transcriptionally induced by ATRA.133,134 CRABPII may sequester ATRA, contributing to therapy resistance.135Tissue glutaminase II, which plays a role in differentiation and apoptosis, is induced rapidly by ATRA136 and contains functional RAREs in its promoter.137 138 

Identification of further direct target genes of RARα relevant to normal and malignant myelopoiesis has accelerated, using techniques such as subtractive cloning,139,140 differential screening,141 differential display (DD),142,143and representative difference analysis (RDA).144 These studies focused on genes induced 24 hours after ATRA treatment and tended to identify indirect targets of ATRA action. These included the IFN-inducible RIG-G145 and the related RI58 gene,146 RIG-E, encoding a GPI-linked cell surface molecule,147 and Jem1,148 a basic/leucine zipper transcription factor gene. In contrast, using RDA, a calcium/calmodulin kinase was isolated from ATRA-treated murine promyelocyte MPRO cells harboring a dominant negative RARα molecule. This gene was activated within a few hours after ATRA treatment in a cycloheximide-resistant fashion and therefore is a reasonable candidate as a direct target of RARα.149 Another report also using the MPRO cell line isolated, by subtractive hybridization, several genes rapidly induced by ATRA treatment.150 One transcript induced threefold by ATRA treatment was identical to that encoding LAPTM5, a lysosomal protein expressed preferentially in hematopoietic cell lines.151 The LAPTM5 promoter contained RAREs, and an LAPTM5-luciferase reporter gene was inducible by retinoic acid. The importance of this gene in differentiation is unknown. Current efforts towards the identification of ATRA target genes are using microarrays of immobilized cDNAs152,153 or oligonucleotides,154 which can monitor expression of thousands of gene simultaneously.155 156Table 2 summarizes some of the salient points regarding myeloid biology, RARs, and APL.

PML structure.

The t(15;17) rearrangement affecting the PML gene on chromosome 15q22 is the molecular basis for approximately 98% of all cases of APL.5 The PML gene locus spans 35 kb and contains 9 exons encoding mRNAs of 4.6, 3.0, and 2.1 kb. Alternative splicing of C-terminal exons yields up to 20 different isoforms of the protein5; however, most cell lines express a similar pattern of isoforms.11-13,157-159 The longest cDNA open reading frame encodes a 560 aa polypeptide with a predicted molecular weight (MW) of 70 kD.10 159 

The PML protein has a modular structure with several domains (Fig 3). These include the following:

Fig. 3.

Functional domains of the PML protein and structure of the PML-RAR and reciprocal RAR-PML proteins generated in t(15;17)-APL. In all patients, the RING finger, B boxes, and at least the first two coiled coil domains of PML are included in the fusion protein. Heterogeneity in the breakpoint within the PML gene leads to a long and short form of PML-RAR depicted, as well as the rarer intermediate form (not shown). The RAR-PML fusion is detected in the majority of cases, but no evident function can be ascertained from its structure.

Fig. 3.

Functional domains of the PML protein and structure of the PML-RAR and reciprocal RAR-PML proteins generated in t(15;17)-APL. In all patients, the RING finger, B boxes, and at least the first two coiled coil domains of PML are included in the fusion protein. Heterogeneity in the breakpoint within the PML gene leads to a long and short form of PML-RAR depicted, as well as the rarer intermediate form (not shown). The RAR-PML fusion is detected in the majority of cases, but no evident function can be ascertained from its structure.

Close modal
Fig. 4.

2D-NMR structure of the PML RING finger.166Spheres representing Zn2+ ions coordinately bind to histidine and cysteine residues allowing loops of protein to extend in a spherical configuration (courtesy of K. Borden).

Fig. 4.

2D-NMR structure of the PML RING finger.166Spheres representing Zn2+ ions coordinately bind to histidine and cysteine residues allowing loops of protein to extend in a spherical configuration (courtesy of K. Borden).

Close modal

(1) A cystein-rich region (aa 57-222) composed of three zinc-finger like structures. The first is a RING (Really Interesting New Gene) finger,160 a Zn2+ binding motif with the configuration C3HC4 (aa 57-91). The following two are called B box zinc fingers (aa 140-161 and aa 189-222).12,13,157,159 The RING finger motif161is found in more than 80 proteins involved in oncogenesis, regulation of gene expression, mRNA processing, and DNA recombination and repair (reviewed previously160-162). RING finger/B Box proteins often are linked to a coiled-coil domain and comprise a subfamily of RING proteins (RBCC for RING-B Box-Coiled-Coil).

The RING finger/B-box region is involved in localization of PML into distinct nuclear domains known as nuclear bodies (NB), presumably through interactions with protein partners (see below).5,163,164 Biophysical studies, including 2-D NMR, showed that the RING finger structure is spherically organized around several loops of protein extending from two Zn2+ ions that coordinately bind cysteine and histidine residues (Fig 4). This positively charged structure precludes DNA binding, implying that the RING domain is required for protein-protein interactions.165,166 PML with mutations in critical RING finger cysteine residues loses its characteristic nuclear body localization and its biological activity as a growth suppressor (see below),164,166-168 but other charged residues on the surface of the globular RING structure may also affect NB formation.169 The B box domains also bind zinc ions and any mutation of the B-box cysteine residues disrupts NB formation.164 Neither the RING finger nor the B-box motifs are required for PML to self-associate, suggesting that interactions with other proteins, through the Cys-rich motifs guide PML into the multi-protein complex of the nuclear body (see below).163,166 170 

(2) A helical coiled-coil region (aa 229-360) consisting of eight heptad repeats with hydrophobic amino acids at the first and fourth positions. This region is responsible for multimerization of PML and heterodimerization with PML-RARα and plays a role in NB localization. This region also interacts with partner proteins and is required for the growth suppression activity of PML as well as the ability of PML-RARα to block differentiation.163,167,170 171 All PML isoforms contain the RING Finger/B Box domains as well as at least the N-terminal coiled-coil motifs.

(3) An N-terminal proline-rich sequence (aa 1-46) that can bind the Arenavirus Z proteins involved in viral genome synthesis172but is not required for growth suppression by PML.167 

(4) A basic sequence, containing a nuclear localization signal (aa 476-490)167,170 required for the biological activity of the protein.167 However, exact nuclear localization in NBs also requires the RING finger/B box and coiled-coil motifs.

(5) An acidic C-terminal Ser/Pro-rich domain of unknown function, highly variable in length due to alternative splicing and rich in potential phosphorylation sites.11-13,157,159 167 

The PML protein.

PML, when expressed after transfection into cells, is detected as a series of 90- to 100-kD protein bands as well as a set of bands in the 70 kD range, as predicted from the amino acid sequence.173-175 Endogenous PML is detected as a 90-kD species along with a variety of other protein species (150-50 kD) due to alternative splicing and covalent modifications.174,176-180 The PML sequence contains potential casein kinase II and proline-directed kinase sites, and32P-labeling studies demonstrated that PML is phosphorylated on serine and to a lesser extent on tyrosine residues.5,170,174,181 Some of the sites are constitutively phosphorylated and others may be cell cycle dependent.182In this regard, PML was found to be a substrate for phosphorylation by Cyclin A/cdk2174 182 in vitro.

PML expression.

PML mRNA was widely expressed in all cell lines tested.5 183 The pattern of PML protein expression in tissues is complex and controversial, possibly due to differences in techniques and antibodies used for immunodetection. In addition, PML mRNA and protein expression are often not concordant, suggesting posttranscriptional regulation. There are a few observations that should be highlighted.

First, PML is highly expressed in inflammatory diseases such as psoriasis and hepatitis, in inflammatory cells surrounding epithelial cancers and Hodgkin’s disease, in inflammatory lesions of graft-versus-host disease, and in activated fibroblasts.183-186 This suggests induction of PML expression by soluble factors, probably IFNs (see below). Second, some, but not all,185 groups found a correlation between the level of PML expression and degree of dysplasia in atypical breast hyperplasia and cervical intraepithelial neoplasia cells. Interestingly, when the breast tumors became invasive, PML expression decreased again.183,184 Some investigators found a correlation between the rate of proliferation of normal tissues and PML expression, with postmitotic cells tending to express higher levels of PML,175,184,185 whereas others187 suggested that PML levels were better correlated with cellular activation by cytokines and rates of protein synthesis. Third, PML delocalization can be associated with neoplasia. Hepatic carcinoma in particular was associated with mislocalization of PML in the cytoplasm rather than the nucleus.183 Fourth, PML and probably other proteins of the nuclear body are induced by hormones such as estrogen and cytokines such as IFN.177,183,184,188,189 Lastly, PML is expressed in myeloid precursors in the bone marrow173,189 and to a lesser extent in circulating monocytes and granulocytes. Activation with IFN leads to reinduction of PML in these cells. This suggests an early role for the protein in myeloid differentiation, perhaps regulating cell growth, and later in host defense.173,175,183,185,190 In lymphoid cells, PML is expressed mainly in postmitotic mature T and B cells and not the germinal center or in proliferating cortical thymocytes.175Hence, PML may play a broad role in mature immune cells.

One of the most striking features of PML is its speckled localization to discrete nuclear domains termed PODs (PML Oncogenic Domains), ND10 (Nuclear domain-10), or NB.170,173,191,192 These structures, originally described more than 35 years ago,193came under new scrutiny when NB proteins, including PML, were detected by human autoimmune antisera.191,194-199 Nuclear bodies vary in both size and number in different cell types. Their presence is roughly proportional to the rate of protein synthesis and inversely proportional to differentiation.187 Strikingly, PML is delocalized from the NBs to a microspeckled nuclear pattern in t(15;17) APL cells and relocalizes to the NB after ATRA treatment.173,191,198 Cells generally contain 10 to 20 doughnut-like or spherical 0.3-0.5-μm NB structures. Electron microscopy showed that, in the NB, PML and other proteins surround an electron dense core that may contain ribonucleic acid.197,198,200 NBs are associated with the nuclear matrix, which plays a role in trafficking of molecules and organization of chromatin within the nucleus. In initial studies, the NBs did not overlap with spliceosomes, centromeres, or sites of RNA transcription.191,197,201 Neither sequence-specific transcription factors such as the glucocorticoid receptor and E2F nor a general factor such as TFIIH were concentrated within the NB,201 which appeared to exclude a transcriptional role for the NB. This notion must be seriously re-examined in light of new data from the lab of Ron Evans, who, using refined techniques, found the presence of nascent mRNA in the center of the NB structure.202 Furthermore, the transcriptional coactivator CBP was found colocalized in the NB with PML. This, together with recent data indicating the presence of PML in the AP1 DNA binding complex,203 suggests that the NB may play a role in stimulating transcription. Whether the NB might be a site of transcriptional initiation, elongation, or processing of the new mRNA transcript is unknown. However, data that PML can repress transcription and the finding of HP1, a heterochromatin component, within the NB204 suggest a role for PML and/or other NB components in downregulation of transcription.

PML-containing nuclear bodies do not colocalize with sites of nascent DNA,191,197 except during mid-S phase, when they are found adjacent to replication sites,201 potentially indicating a role for the NB in this process. Although PML-containing nuclear bodies are distinct from the coiled body, involved in mRNA splicing,205 the two structures were often found adjacent to each other, suggesting a functional interrelationship.201 PML was also found in the interchromatin granules, which are thought to be sites of RNA splicing,206 and can be found in the nucleoplasm in a diffuse staining pattern192 as well as in a cytoplasmic granular pattern.175,189 Although the exact site and mechanism of PML action is still uncertain, nuclear expression of PML seems to be essential for its biological function, because PML mutants lacking the NLS were unable to suppress oncogenic transformation.167 However, certain spliced isoforms of PML, devoid of the C-terminal NLS, are found exclusively in the cytoplasm,175 but other isoforms that do contain the NLS can be found in both the nucleus and cytoplasm of transfected cells. Hence, a cytoplasmic role for the protein is not ruled out and neither is the possibility that PML may shuttle between the nucleus and cytoplasm. The partition of PML between the nucleoplasm and the NBs may be controlled by processes such as differential phosphorylation and conjugation of PML to a ubiquitin-like molecule, sentrin.179 

The presence of the RING finger, B box motifs, and coiled-coil motifs are all necessary for PML to properly localize in the NBs.167 Mutants lacking these structures sequestered normal PML from the NBs in a dominant negative manner.167,175 In addition, forms of PML with mutations in critical cysteine residues required for normal protein folding failed to localize in the NBs.164,166,170 PML expression and NB structure are dynamic. Augmenting the cellular levels of PML either by transfection with a PML expression vector or by treating cells with IFNs increased the size and number of nuclear bodies, possibly due to deposition of PML and recruitment of other proteins to these sites.177,189,192,198,207 In contrast, PML-RARα caused disappearance of nuclear bodies from the nucleus as determined by staining with antibodies directed against PML and the SP100 NB protein.198 

The pattern and intensity of PML expression changes across the cell cycle. Cells in G0 exhibit few nuclear bodies and show weak staining for PML. As cells are stimulated into the cell cycle, the number of NBs and their intensity of staining with PML antibodies increases. As cells progress through S to G2 phase, PML disperses to multiple smaller dots and gradually fades, with two or three of these residual structures left in mitosis.174,183,184 Prolonged amino acid starvation208 or induction of cell senescence209 induces the coalescence of the NBs into 2 to 3 large structures. When amino acid-starved cells were rescued by addition of fresh nutrients, the normal pattern of 10 to 20 smaller NBs reappeared.208 The pattern of PML expression also responds to cell stresses such as heat shock,192γ-irradiation,200 and viral infection (see below). How these changes relate to the function of the nuclear body is unknown.

Findings related to transcriptional properties of PML have been contradictory but intriguing. PML specifically suppressed transcription of the MDR and EGF-receptor (EGF-R) promoters.210 However, PML enhanced transcription of the CD18 promoter210 as well as the promoter for the gene encoding the major histocompatibility complex (MHC) class I transporter molecule TAP-1.211 Furthermore, PML increased the transcriptional activity of the progesterone receptor (PR) in the presence of hormone up to 20-fold and also stimulated transcription mediated by the mineralocorticoid, glucocorticoid (GR), and androgen receptors, but not the RARs.212 Deletion analyses indicated that both activation domains (AF1 and AF2) of PR as well as the Cys-rich and coiled-coil domains of PML were required for the PML effect on transactivation. PML did not increase the affinity of PR for progesterone, increase DNA binding by PR, or coprecipitate with PR. This does not preclude PML as a transcriptional cofactor or adaptor to the basal transcriptional machinery. Alternatively, PML might sequester a negative regulatory factor from the PR, indirectly stimulating its activity. The physiological relevance of this interaction must be tempered by a report that PML does not colocalize with the GR.201 

GAL4-PML fusions were found to repress a reporter gene containing GAL4 binding sites,213,214 although an earlier publication did not note such an effect.170 The repression domain mapped to the coiled-coil region and the C-terminal serine-rich region but not to the RING finger; the magnitude of repression was cell-type and promoter specific. Mutational analysis indicated that the coiled-coil, not the RING finger motif was required for repression and that localization to the NB was not required. Rather, the nucleoplasmic fraction of PML may be responsible for this effect. It is tempting to speculate that PML, like PLZF (see below), interacts with histone deacetylase corepressors to repress transcription. Another mechanism of action of PML was suggested by the finding that PML-mediated repression of the EGF-R promoter was mediated through Sp1 sites. PML was then found to inhibit the DNA binding by Sp1 protein.215 These data suggest that, under certain circumstances and probably not when localized to the nuclear body, the PML protein could modulate gene expression by direct interaction with specific transcription factors. This function might be abrogated by PML-RARα, which, when coexpressed with Gal4-PML, leads to simulation of transcription rather than repression.214Although PML might be a transcriptional corepressor, a recent report also indicated that PML can stimulate transcription mediated by fos and jun through AP1 sites.203 

The matrix association of PML makes it difficult to study the interactions of PML with putative partners, because aggressive purification procedures required to extract PML from the nuclear matrix can disrupt protein-protein interactions.216 It is interesting to note that two other RARα translocation partners, NPM and, in particular, NuMA, are also closely associated with the nuclear matrix. In addition, the remaining fusion partner, PLZF, is also found in (the matrix-associated) nuclear bodies. Together, these observations indicate that disruption of nuclear matrix function may be a common theme in APL pathogenesis, perhaps further disturbing normal patterns of gene expression.

Nearly 20 natural components of the nuclear body have been identified to date (Table 3). The diverse nature of these proteins and the fact that they often localize both within the nuclear body and other subnuclear patterns suggests that the NBs may not be a site of active cellular metabolism but rather a storage site for nuclear proteins whose temporal and spatial expression must be tightly controlled. However, several of these proteins are induced by IFN, including PML, SP100, and PLZF,216-219 pointing to an alternative hypothesis that the nuclear bodies may play a role in cellular proliferation and/or the antiviral response.

Only a few proteins are known to actually bind directly to PML. The first of these, sentrin, originally called PIC1 (also called SUMO-1 and UBL-1), was identified in a yeast two hybrid screen.220Both cotransfected and endogenous PML and sentrin colocalized in the NB with variable overlap. In the APL NB4 cell line, sentrin was partially delocalized to PML-RARα microspeckles. Sentrin is a ubiquitously expressed 11.5-kD peptide containing a ubiquitin homology (UbH) domain.220 Two other groups independently cloned sentrin by its association with the rad51 DNA repair protein221 and the ranGAP protein of the nuclear pore complex,222 whereas a third group found that sentrin interacted the tumor necrosis factor (TNF) receptor. This group found that sentrin covalently attaches to proteins in a manner akin to ubiquitin, suggesting that sentrin may play a role in protein turnover.223 Furthermore, this group found that overexpression of sentrin protected cells from fas/apo- and TNF-induced apoptosis.224 Given the role of PML as a growth suppressor (see below), it is possible that PML sequesters sentrin, promoting apoptosis and balancing the function of sentrin. However, it is more likely that sentrin modifies the function of PML and other components of the nucleus. Newer data show that sentrin covalently binds to PML in the RING finger motif, the B box domain, and the more C-terminal nuclear localization signal of PML.178,180,225,226 This association was exclusively found in nuclear localized PML, particularly with PML associated with the NBs,225,226 suggesting that the sentrin modification targets PML to the NB. However, sentrin was not associated with PML in mitotic cells, suggesting that the modification is cell cycle dependent. It was reported that PML-RARα is not modified by sentrin180,226 and that sentrinization was not restored to this protein after ATRA treatment. This was surprising given the moieties modified by sentrin are present in PML-RARα, and another group found that, when expressed in U937, PML-RARα but not PLZF-RARα became conjugated to sentrin, suggesting that the molecule might play a role in the degradation of PML-RARα after ATRA treatment.227 

The PLZF protein may be a key interaction partner of PML. The PLZF protein, originally characterized by its fusion to RARα in t(11;17)(q23;q21)-associated APL, colocalizes with PML both in myeloid and in transfected nonhematopoietic cells.216Colocalization was incomplete, and one group estimated only approximately 30% overlap of PML and PLZF in KG1 cells.228The PLZF nuclear dots and PML NBs also appeared to be in different functional compartments, because expression of the adenovirus E4 protein in KG1 cells delocalized PML from the NB (see below) but did not affect the expression pattern of PLZF. In semistable transfected cells, several days were required for the PML and PLZF proteins to sort within the cell and become colocalized, suggesting complex regulation of this association.216 Nevertheless, interaction between PML and PLZF was demonstrated by biochemical assay, and the association was found to be mediated by the coiled-coil domain of PML and the first two zinc fingers of PLZF (A. Zelent, personal communication, December 1998). PLZF could be delocalized to a microspeckled pattern in NB4 cells and reverted to the NBs upon treatment with retinoic acid. These provocative findings suggest that PML may be involved in the transcriptional modulation mediated by PLZF or, conversely, that PML regulates availability of PLZF by sequestration within the NBs.216 Thus, a common mode of leukemogenesis may exist in APL, based on disruption of a pathway that contains both PLZF and PML.

New data indicate that the retinoblastoma (Rb) protein interacts with PML. PML could be coimmunoprecipitated with a low percentage of the underphosphorylated form of Rb but not the related p107 and p130 proteins. As seen by confocal microscopy, the speckled fraction of Rb in the cell overlapped the PML NB. PML-RB interaction required the pocket domain of Rb and the first two coiled-coil motifs of PML, with the RING finger motif, stabilizing the complex. In addition, a PML isoform with an extended serine-rich C-terminal domain bound significantly less strongly to Rb, suggesting that different PML isoforms could differentially bind Rb.229 PML-RARα also bound to Rb and delocalized Rb from its usual speckled pattern into the microspeckled pattern characteristic of PML in APL. The functional consequences of this interaction in growth control are uncertain, because both PML and PML-RARα are able to inhibit the growth of nonhematopoietic cells devoid of functional Rb. However, it remains fascinating to postulate that one of the mechanisms through which PML inhibits cell growth is by limiting accessibility of Rb to the cyclin-dependent kinases that phosphorylate it during G1 phase.

Finally, a novel function for PML was suggested by its association with the ribosomal P proteins P0, P1, and P2.230 These proteins form part of the large ribosomal subunit, are localized in both the nucleus and cytoplasm, and are involved in the process of translation,231-233 suggesting that PML may have a role in translational control. This is supported by the findings that PML interacts with the L7 leucine zipper protein and EF-1,220also implicated in translation, and that PML partially localizes to the cytoplasm.175 It is intriguing to note that the NPM protein, fused to RARα in t(5;17) APL,234 is involved in ribosome biogenesis and shuttling ribonucleoproteins between the nucleus and cytoplasm.235-237 This places both PML and NPM in a similar functional axis.

PML is an IFN-responsive gene that may be important for its normal immunity. In hematopoietic NB4 and nonhematopoietic cell lines, PML and PML-RARα mRNA were induced fivefold to 30-fold in response to IFN in a dose-dependent manner.177,188,189 Actinomycin D blocked this effect, whereas cycloheximide did not, indicating that PML is a primary target of IFN action. Other cytokines that induce the JAK/STAT pathway did not cause this increase. The PML promoter contains an ISRE (IFN-stimulated response element) as well as a GAS (γ activation site), both of which bind to STAT proteins.158 After IFN treatment, different PML isoforms become visible with increases in the intensity of staining and number of nuclear bodies. STATs are involved in upregulating multiple components of the NB, including Sp100, whose promoter also contains a GAS and ISRE.176 Because PML seems to be a growth suppressor, it may be an attractive target for the antiproliferative and antiviral effects of IFN.158 Finally, ATRA can cooperate with IFN to synergistically induce PML expression through induction and activation of STATs acting through the GAS element in the PML promoter.118 Induction of wild-type PML could thus play a role in the therapeutic effect of ATRA in APL.

It is apparent that one of the earliest events of both lytic and latent viral infections is the targeting of viral products to NBs, often resulting in the reorganization of PML localization and NB architecture. Some of these results are summarized.

Adenovirus.

Adenovirus infection causes PML to undergo dynamic spatial redistribution. As the virus progresses through its life cycle, the NBs disappear and become part of the viral nuclear inclusions.206 Mutational analysis showed that the adenovirus E4-ORF3 gene product was responsible for NB redistribution to fibrous structures or tracks. Overexpression of PML blocked this redistribution, an effect that was lost when PML deleted for the coiled-coil domain was used.238 

HSV-1.

ICP0, an immediate early gene, encodes a RING finger protein that is required for both the lytic and latent herpes virus life cycles. ICP0 activates viral gene expression, targets the NBs, colocalizes with PML, and modifies NB architecture.239 HSV-1 replication sites, with attachment of viral DNA to the nuclear matrix, appear to coincide with NBs as well,240 suggesting that NB components may be involved in viral DNA replication. In latent infections, in which HSV genes are partially expressed from episomes, viral genomes may also be associated with NBs. Stress, which disperses the NBs, also allows the virus to come out of latency.240 

Epstein-Barr virus (EBV).

In contrast to the viruses listed above, EBV tends to transform lymphocytes by latent infection rather than lysing target cells. EBNA 5 is an early protein critical for transformation and latency. This protein is colocalized with PML in interphase NBs, with PML coating the outside of the structure and EBNA found within.241 242 In contrast to the other viral infections, the NB structure is not disrupted by EBV infection.

Arenavirus (LCMV, Lhassa).

The arenavirus RING finger containing Z protein binds to the N-terminal proline-rich region of PML, resulting in its delocalization to the cytoplasm.172 The fact that the Z protein targets P ribosomal proteins that are partners of PML suggests viral hijacking of the cellular translational machinery, of which PML may be a component.230 Alternatively, PML may be moved to the cytoplasm to prevent it from promoting apoptosis (see below), thus enabling survival of virus-laden cells and allowing the virus to establish chronic infections.168,230 This list is by no means complete, because cytomegalovirus (CMV)243,244 and papilloma virus245 also target the NBs.

To test whether PML plays a role in the antiviral effect of type I IFN, PML−/− embryonic fibroblasts were infected with a number of DNA viruses, including herpes simplex and vesicular stomatitis virus, in the presence or absence of IFN.207 There was no difference in the efficacy of IFN in decreasing resultant viral titers; therefore, the absence of PML did not confer increased susceptibility to viruses. Engineered expression of PML in 3T3 cells did not confer resistance to HSV, VSV, EMCV, or Adenovirus.207 However, another group found that in HepG2 cells, overexpression of PML inhibited adenovirus growth, perhaps by delaying or blocking NB protein recruitment to viral replication domains.238 A third group found that PML overexpression selectively inhibited replication of influenza virus but not of encephalomyelitis virus. In contrast, overexpression of the Sp100 NB protein had no significant antiviral effect.246 Together, it can be concluded that, although PML itself is not absolutely required for the activity of IFN, it may confer some of the antiviral activity of IFN.

These findings suggest that NBs could be carefully controlled organelles that store regulatory factors involved in viral transcription or replication.239 Alternatively, NBs may be an intranuclear defense system. This notion is supported by the localization of infecting virus genomes to the vicinity of the NB, the IFN induction of NB proteins, and the growth-suppressive properties of PML.240 Perhaps more plausibly, viruses may target and disrupt the nuclear body to abrogate an apoptotic program (see below), allowing the virus to promulgate freely in the cell population. The delocalization of NB proteins caused by PML-RARα expression could mimic activation of the viral program, possibly resulting in uncontrolled cellular proliferation.238 

The growth-suppressive actions of PML were suggested by the finding that NB4 cells selected for expression of exogenous PML harbored mutations in PML, suggesting that the wild-type protein was not tolerated.210,247 Infection of NB4 cells with a retrovirus harboring PML suppressed the ability of the cells to form colonies in soft agar. In addition, conditioned medium from these cells suppressed colony formation of wild-type NB4 cells, suggesting the release of negative growth control factors.210 Furthermore, PML-overexpressing NB4 cells, when injected into nude mice, yielded smaller tumors that appeared with a longer latency than vector-expressing cells.210 Attempts to circumvent the toxic effect of PML led another group to create an episomal transfection system in which it was demonstrated that even low levels of PML were toxic to NB4 cells. Because RARα overexpression was also growth suppressive to these cells, these investigators concluded that, in t(15;17) APL, the reduced dosage of PML and RARα could contribute to uninhibited cell growth.247 

It was further shown that PML inhibited transformation of rat embryo fibroblasts expressing Ha-ras and mutant p53 or Ha-ras and c-myc.210 However, PML did not inhibit all oncogenic pathways. PML suppressed foci formation in NIH 3T3 cells transformed by the neu oncogene, but not by the ras oncogene. PML-RARα acted in a dominant negative manner to prevent suppression of transformation by PML, sequestering PML in the cytoplasm, whereas PML-RARα itself did not stimulate the formation of foci. The neu oncogene-transformed NIH-3T3 cells mentioned above were reverted by retrovirus-mediated transfer of PML, resulting in restitution of wild-type morphology, contact inhibition, and anchorage-dependent growth. There was no major difference in cell cycle distribution of PML-expressing cells, suggesting that PML suppressed growth not by cell cycle inhibition but by altering cell survival and apoptosis. Although PML expression led to decreased expression of the neu protein, the cell cycle profile and morphologic response of cells to expression of PML differed from that of cells treated with an anti-neu antibody, which arrest in the G2 phase of the cell cycle. This suggests that PML interferes with downstream targets of the neu signal transduction molecule, perhaps reflective of the normal physiologic function of PML.248 

However, in another system, PML did affect the progression of the cell cycle. When overexpressed in HeLa cells, PML inhibited cell growth, colony formation in agar, and tumor growth in nude mice, as in the models described above, but also caused an accumulation of cells in G1249 and a delay of cell entry into S phase, correlated with decreased expression of cyclin E and cdk2. Similarly, expression of PML in breast cancer cell lines blocked cells in G1associated with a decrease in cyclin D1 and CDK2 and accumulation of hypophosphorylated Rb.250 This is intriguing given recent data that hypophosphorylated Rb and PML can complex.229However, PML can suppress growth even of Rb-defecient cells, suggesting that complexes with other Rb-like proteins could play a role as well.

de Thé’s group stably expressed PML in HeLa, CHO, and NIH 3T3 cells and observed a twofold to fivefold decrease in growth rate, decreased colony formation, and inhibition of tumor formation in nude mice.184 Growth suppression by PML was further augmented by IFN, perhaps by stimulating expression of other nuclear body proteins.184 A structure/function analysis of the PML protein indicated that deletion or mutation of the RING finger motif of PML abrogates both NB formation and growth suppression.166-168,170 Deletion of the coiled-coil motif yielded a diffuse nuclear pattern of expression and no growth suppression, whereas deletion of the NLS of PML or expression of a splice variant of PML leads to a granular cytoplasmic pattern of expression and also abolished growth suppression.251 The proline-rich region at the N-terminus of PML and the serine proline variable region at the C-terminus of the protein were dispensable both for NB formation and growth suppression.167 Thus, PML expression in the nucleus is required for growth suppression, although a recent report questions whether expression in the NB per se was required for growth suppression.251 In support of this observation, infection of NIH 3T3 cells with LCMV virus rapidly relocalized PML into the cytoplasm and delayed apoptotsis after serum starvation.168 Similarly, treatment of cells with antisense PML oligonucleotides delayed death after serum starvation. These results suggest that the correct localization of PML plays an important role in apoptosis and cell growth.

Recent data suggest that PML may promote cell death by novel mechanisms. PML interaction with the P0 ribosomal protein230 could target the 28S rRNA for cleavage, an important step during apoptosis.230 PML expression yields cell death that is not associated with the usual chromatin condensation or activation of caspase 3 the major effector of apoptosis.252 Paradoxically, zVAD a caspase inhibitor accentuated PML-mediated apoptosis and increased PML expression levels in the NB, as did arsenic (see below). PML was found to recruit the pro-apoptotic BAX protein to the NB as well as the cell cycle inhibitor p27KIP1; the importance of this recruitment remains unclear, because it is still not certain what processes occur in the NB. Use of PML−/− animals yielded somewhat different results, with a 50% decrease in apoptosis of T cells after γ-irradition and a great reduction in fas-induced apoptosis.253 PML null mice had no significant difference in expression of multiple apoptosis mediators, with the exception of caspase 1 and 3, whose activities, required for radiation and fas-mediated apoptosis, were markedly reduced. Furthermore, PML null animals were resistant to ceramide, TNF, and IFN-mediated apoptosis. Together, these data indicate a critical role for PML in multiple apopotic pathways. High-level expression of PML may induce apoptosis in the absence of caspase action, whereas physiological levels may be required for the normal activation of caspases.254 

It is certain that PML is growth suppressive when overexpressed, but whether it is a tumor suppressor is uncertain. Mutations of PML have not been reported in other forms of cancer; even in t(15;17) APL, patients would not be expected to be null for this protein. However, new data from experiments with targeted disruption of the PML gene support a tumor-suppressor function for the protein.132,190PML−/− mice were viable and fertile but highly susceptible to fungal or bacterial infections despite a strong inflammatory response,132 suggesting a functional defect in the inflammatory cells. Circulating and bone marrow mature myeloid cell counts were modestly decreased in these animals, indicating that, although PML was not required for the formation of the myeloid lineage, it may be involved in efficient terminal differentiation of these cells. Early death of these mice from infection prevented the long-term follow-up required to detect spontaneous tumor formation. However, the skin of PML−/− animals treated with the chemical tumor initiator DMBA formed twice as many papillomas as that of wild-type animals. In addition, animals treated with DMBA systemically formed twice the number of lymphomas as PML replete animals. PML null murine embryo fibroblasts proliferated more rapidly than wild-type fibroblasts and more readily formed colonies in soft agar. Whereas ATRA suppressed the growth of wild-type fibroblasts, there was little effect on the PML−/− cells. Whereas IFN inhibited the proliferation of normal marrow precursors as determined by clonogenic assays, α-IFN or γ-IFN had no myelosuppressive effect on PML−/− marrow.132 

PML may inhibit tumor growth in other ways as well. In a remarkable new study, PML was found to stimulate expression of MHC class I antigens and the transporters responsible for moving peptides to the cell surface in association with class I antigens.211 Hence, disruption of PML expression, whether by tumor viruses such as adenovirus or by the PML-RARα fusion protein, might lead to decreased presentation of viral or oncoprotein antigens and defective immune surveillance for tumors. A recent study implicates PML in growth control by p53. PML expression was induced fivefold to 10-fold at the posttranscriptional level by ionizing radiation255 or a DNA-damaging agent. Overexpression of p53 in HeLa cells was also correlated with PML induction and G1 arrest, suggesting that PML might be considered a GADD (growth arrest and DNA damage) gene.256 

These exciting data suggest that PML can inhibit cell growth and may be required to mediate some of the physiological actions of the IFNs. To study potential effects of PML on RARα signaling, an important target gene of RARα in differentiation, p21WAF1/CIP1, encoding a cyclin-dependent kinase inhibitor, was studied. Interestingly, p21 WAF1/CIP could not be upregulated in the PML−/− fibroblasts. Thus, PML might be required for certain pathways of retinoid signaling. Therefore, PML-RARα might also disrupt RARα function in APL by blocking the ability of wild-type PML to cooperate with RARα to stimulate myeloid differentiation. The L7 leucine zipper protein, a PML partner,220 modulates the transcriptional activity of nuclear receptor signaling complexes.257 258 It is therefore conceivable that this might mediate the effects of PML on retained nuclear receptor function.

In view of the emerging evidence, it is reasonable to describe PML as a tumor-suppressor protein involved in the growth suppression, differentiation, and immune response pathways of certain cytokines such as IFN. The mechanism by which PML encourages growth arrest is unclear, although interaction with Rb and related proteins offers an intriguing avenue. PML induces apoptosis in the absence of new protein synthesis252 and may act in both a caspase-dependent and -independent manner,254 suggesting that its role in transcription may be secondary to its role in growth control. Apoptosis by PML is intimately related to its localization in the NB organelle. The NB may be a repository for growth and apoptosis regulators, released or sequestered according to environmental cues, and could be a key component of the cell’s defense system against viral infections. Delocalization of PML by PML-RARα may be a critical step in the pathogenesis of APL. Supporting this notion, PML-RARα inibited fas-mediated suppression of myeloid growth. When expressed in the PML+/− background, PML-RARα further inhibited apoptosis, suggesting that PML-RARα works in part by subverting normal PML function.253 In opposition to this view, the other forms of APL occur without removal of PML from the NB, and differentiation block by PML-RARα can occur without delocalization of PML. If the PML-RARα fusion is a novel gain of function mutant and/or makes a critical interaction with another NB protein also in an apoptotic pathway, then the presence or absence of PML becomes irrelevant to leukemogenesis. A balanced view suggests that PML-RARα itself is leukemogenic, but the tumor-suppressor function of wild-type PML acts to oppose this effect. This is supported by recent experiments crossing PML-RARα transgenic and PML null mice. APL could indeed develop very rapidly in the PML null background, more slowly in PML−/+ mice, and with the longest latency in PML+/+ mice.259 

Structure.

Translocation (15;17) fuses the RARα and PML genes and generates a PML-RARα fusion transcript (Fig 3). Cases of APL that have morphologically normal chromosomes or other cytogenetic abnormalities usually have cryptic rearrangements of PML and RARα.12,260-264 PML-RARα cDNA was cloned from libraries derived from leukemic blasts of APL patients.11,13,15,157,170 Comparison of the cDNA structures obtained by multiple groups showed variation in the amount of PML sequences included in the fusion protein. The RARα portion was invariant, containing the DNA-binding and ligand-binding motifs (B-F domains).265 The PML sequence variation seen among patients was generated by heterogeneous breakpoint cluster regions as well as by alternative splicing.265-269 The most 5′ breakpoint,bcr3, fuses PML exons 1 through 3 of PML, encoding the RING, B-boxes, and coiled/coil domains to RARα exon 3, encoding the B domain of the receptor. This breakpoint yields short PML-RARα fusion proteins [PML(S)-RARα]. Bcr1 is more 3′ within the PML gene and includes sequences from PML exons 5 and 6. This fuses up to 554 amino acids of PML to RARα and has C terminal serine-rich sequences of PML that are putative sites of phosphorylation [PML(L)-RARα]. Breakage in bcr2 involves sites in and around exon 6 of PML and leads to an intermediate length of PML sequence [PML(V)-RARα]. In general, 70% of patients exhibit PML(L)-RARα, 20% PML(S)-RARα, and 10% PML(V)-RARα,270,271 with PML(S)-RARα and PML(L)-RARα representing extremes of contiguous PML sequence fused to RARα. Internal splicing of portions of PML exon 3 led in one patient to a small PML-RARα fusion protein that contained the RING fingers, B boxes, and the first two portions of the α-helical coiled-coil domain, representing the minimal PML moiety required for oncogenicity.265 Each t(15;17) APL patient exhibits a unique set of PML-RARα fusion products indicative of a single breakpoint with alternative splicing, highlighting the clonal nature of the disease. Detection of the PML-RARα fusion transcript by reverse transcription-polymerase chain reaction (RT-PCR) is a sensitive272 and specific test for the diagnosis of APL and can be used to measure minimal residual disease after chemotherapy, differentiation therapy, and bone marrow transplantation.273,274 Reappearance of PML-RARα transcripts in the marrow often precedes a frank leukemic relapse.5,275-277 Initial studies indicated that patients treated with ATRA who harbored the PML(S)-RARα had a high likelihood of early death or relapse.278,279 One in vitro study indicated that blasts from APL patients with the PML(V)-RARα isoform had decreased ATRA sensitivity.280 There was an association between the PML(S)-RARα isoform and more primative morphology281 and secondary cytogenetic abnormalities, suggesting a biological difference between the isoforms,271possibly due to abnormalities of DNA repair or cell cycle control. Despite the potential differences among the PML-RARα isoforms, numerous studies reported consistently good clinical outcomes in all APL patients,271,282 283 probably due to the highly effective nature of current therapy.

The mechanism by which the t(15;17) translocation occurs is not known. It may be that many illegitimate recombinations occur during normal cell division and are eliminated by DNA repair systems.284,285 Recent analysis found short stretches of identity between the PML and RARα genes in the breakpoint regions.286 It was proposed that random cleavage of the RARα and PML genes is followed by limited pairing of short stretches of homologous DNA, repair of the breaks, and joining of the loci. Those clones that contain the PML-RARα transcript survive and have a growth advantage.

The PML(L)-RARα fusion transcript yields proteins of 110 and 120 kD and PML(S)-RARα species of 103 and 90 kD,170,174,287possibly resulting from alternative start codons.11,13,15,157,170 The fusion proteins of transfected cells or NB4 cells are about 10 kD larger than proteins generated by in vitro translation, suggesting the presence of posttranslational modifications.170,287 In APL cells, PML-RARα is present in great excess over wild-type RARα, making it the predominant retinoid receptor in those cells.265 287 

Protein-protein interactions.

PML-RARα oncoprotein, an aberrant retinoid receptor with altered DNA binding activity,11,13,15,157,170,171 can bind RAREs as a homodimer,171,287 whereas wild-type RARα cannot.288 Homodimerization requires the coiled-coil domain of PML and not the E/F ligand binding/dimerization moiety of RARα (Fig 3). The first 2 (of the 4) hydrophobic clusters of the coiled-coil region also mediate PML-RARα/PML association. The smallest PML-RARα protein identified contained only clusters 1 and 2 (Fig3).265 PML-RARα homodimers can be detected as a distinct DNA-binding species in nuclear extracts from NB4 cells287and display weaker affinity for certain artificial and natural RARE sites than RAR/RXR heterodimers.287 When combined with RXR, PML-RARα forms multimeric complexes on the RARE,171,287,289 and even a 1:1 molar ratio of in vitro translated RXR to PML-RARα favors the formation of PML-RARα/RXR heterodimers.171,289 Hence, the existence of the PML-RARα homodimer complex in NB4 extracts probably reflects the high level of expression of PML-RARα relative to wild-type RARα and RXR in APL cells. When bound to RAREs along with RXR, PML-RARα displays the same binding site preference as wild-type RARα. The PML-RARα/RXR interaction does not require the DNA binding domain of the RARα moiety within PML-RARα,171 but can occur through the E/F domain of RARα. Multimeric complexes from transfected cells may reflect the ability of PML-RARα/RXR heteromers on one DNA binding site to associate through the PML coiled-coil domain with heteromers on other sites. In the cell, this could reflect the ability of PML-RARα to efficiently bind to RXR and sequester it from wild-type RARα. These multimeric PML-RARα/RXR complexes were also seen on EMSA in extracts derived from NB4 cells.287 In addition, PML-RARα, detected by size exclusion chromatography by its ability to bind [3H]-ATRA, elutes with an apparent MW of 600 to 1,300 kD, further supporting the idea that PML-RARα multimerizes with itself and/or other proteins.290,291 Reinforcing these studies, confocal microscopy showed that PML-RARα draws RXR from its usual subnuclear localization into the compartment occupied by PML-RARα.170 

Taken together, these data suggest that PML-RARα may affect ATRA-mediated signaling through several mechanisms: (1) binding of PML-RARα homodimers to a novel set of target genes, (2) binding of PML-RARα as homodimer or heterodimer with RXR to RARα target genes in competition with RARα, and (3) high levels of PML-RARα in APL cells could sequester RXR and/or other RARα cofactors in a novel nuclear and/or cytoplasmic compartment.

The PML-RARα protein has altered transcriptional properties. Many groups observed that, in the absence of ATRA, PML-RARα represses transcription from RAREs to a greater extent than RARα.13,170,292 This may be the most important quality of PML-RARα. There were conflicting reports regarding transcriptional activation by PML-RARα. In some reports, both PML-RARα (S) and (L) stimulated ATRA-mediated transactivation more strongly than RARα,13,15 whereas others found that PML-RARα activated weakly or not at all.170 Some of these differences may have been due to the use of different cell types or reporter genes. In general, when coexpressed with RARα, the fusion proteins behaved in a dominant negative fashion, reducing activation to the level of PML-RARα alone.157,292 RARα and PML-RARα, although activating transcription to different extents, had a similar ED50 for ATRA (∼5 × 10−9 mol/L) and had similar dissociation constants for ATRA (∼10−10mol/L).170,290 Hence, the altered transcriptional activity of the PML-RARα fusion protein is not related to impaired ability to bind ATRA. However, there are some subtle differences among the PML-RARα isoforms in terms of ATRA binding. The PML(S)-RARα isoform had a higher affinity for 9-cis retinoic acid than PML(L)-RARα.291 A functional consequence of this effect was that PML(S)-RARα activated a reporter gene at lower concentrations of 9-cis retinoic acid than the long isoform. Furthermore, 9-cis retinoic acid was found to be more effective than ATRA in inducing differentiation of APL cells harboring the short isoform in vitro. These data strongly support the notion that differences in clinical presentation of APL are a consequence of PML-RARα variants.

The impaired ability of the PML-RARα protein to activate certain promoters is not due to deletion of the A (activation) domain of RARα. In parallel transfection experiments, a ΔA-RARα mutant activated transcription to a similar extent as wild-type RARα.170,292 However, nuclear receptor A/B domains are known to act in a cell-type specific manner, and deletion of this moiety could still explain some aspects of PML-RARα function.42 Impaired activation is probably due to the ability of PML-RARα to bind the corepressors SMRT and N-CoR more tightly than wild-type RARα, requiring pharmacologic doses of ATRA (10−6 mol/L) for disassociation. This is in marked contrast to the physiologic heterodimer of RARα-RXR, which releases corepressors at 10−9 mol/L ATRA, potentially explaining the need for high doses of ATRA to stimulate differentiation in APL.293-296 The chimeric protein had increased affinity for SMRT and N-CoR, despite the fact that neither corepressor bound to PML in vitro or colocalized in vivo, suggesting that fusion of PML to RARα might modify the E (ligand and corepressor binding) domain of RARα in an allosteric manner. Underlining the central role of the E domain of RARα, PML-RARα mutated in the ligand binding site was unable to release SMRT, even in the presence of large amounts of ATRA.295 

The corepressors N-CoR and SMRT are part of a multiprotein complex that includes histone deacetylases (HDACs). Deacetylation of histones alters the conformation of chromatin and its accessibility to the transcriptional machinery, resulting in transcriptional silencing.46 Binding of ligand to RARα leads to the release of corepressors and binding of coactivators, many of which may acetylate histones. However, PML-RARα may retain the deacetelyase complex, moderating the ability of the protein to activate transcription. In fact, PML-RARα associates with HDAC1 (histone deacetylase 1),295,296 and this may be the basis of the block of critical myeloid gene expression at physiological doses of ATRA. The association of PML-RARα with N-CoR, SMRT, and HDAC1 was further characterized by demonstrating that a fusion protein mutated in the CoR-box (corepressor binding motif) was both unable to associate with HDAC1 and block differentiation of transfected cells.296 Sodium butyrate297 or trichostatin A (TSA),298 inhibitors of histone deacetelyation, stimulated ATRA-mediated transcription by PML-RARα,294-296,299possibly by blocking the activity of residual bound deacetylase complexes. This could explain the synergism of sodium butyrate and ATRA to accelerate differentiation of APL.294,299,300 In fact, when exposed to TSA, a resistant NB4 cell line containing PML-RARα with the E domain mutation mentioned above was able to partially differentiate in response to ATRA,295 presumably by augmenting the function of endogenous wild-type RARα. TSA was able to restore ATRA-induced transactivation of an RARE-containing reporter in these same resistant cells and could potentiate differentiation and expression of RARα target genes in transfected U937 cells.295 296 

PML-RARα also affects other transcriptional pathways important for myeloid differentiation. ATRA and RARα can inhibit transcriptional activation by the AP1 protein, possibly by a competitive effect between the ligand-engaged receptor and either fos or jun for a limiting amount of a common coactivator protein p300 and/or CBP.59,73Somewhat paradoxically, PML-RARα, when coexpressed with fos and jun, stimulates transcription from an AP1 binding site containing reporter gene in the presence of ATRA.301 Whereas PML could not be shown to directly interact with fos or jun, PML could be detected in an AP1 DNA-protein complex.203 

PML-RARα inhibits transcription by other nuclear receptors. In EMSA assays, PML-RARα competes with VDR for RXR and prevents formation of the VDR/RXR DNA-protein complex. In cotransfections assays, PML-RARα blocked vitamin D3-mediated transcriptional activation, an effect reversed by the overexpression of RXR.171 PML-RARα also blocked ligand-mediated activation by the peroxisome proliferator,287 likely by a similar mechanism of sequestration of coactivators. In contrast, PML-RARα, like PML, stimulated transcription mediated by the PR.212 The mechanism of this activity is obscure, but this finding underscores the pleiotropic effects of PML-RARα. Lastly, whereas RARα and STAT1α synergistically stimulated transcription from an IFN response element-containing reporter, PML-RARα did not, suggesting that the cross-talk between IFN and retinoid signaling may be defective in APL.120 

Distillation of these studies shows several points. (1) The transcriptional activity of PML-RARα varies depending on cell type and target promoter. (2) PML-RARα tends to suppress transcription of RARα target genes at physiological concentrations of ATRA to a greater extent than wild-type RARα. This effect, due to aberrrant interactions with corepressors, may be critical for differentiation block in APL. (3) On some promoters, PML-RARα can activate transcription to a similar or greater extent than wild-type RARα at 10−6 mol/L ATRA. (4) PML-RARα can suppress ATRA-mediated transcription by endogenous RARs as well as transfected wild-type receptors. This may be due to competitive binding by PML-RARα, which on some promoters displays intrisically lowertrans-activation potential, as well as competition with RARα for coactivators. (5) The synergistic effects of butyrate or TSA and ATRA on transcription by PML-RARα and in differentiation are likely due to inhibition of histone deaceteylation and alterations of chromatin stimualting activation of RARα targets. (6) PML-RARα also effects the transcriptional function of other nuclear receptors as well as other transcription factors such as AP1 and STATs.

ATRA-resistant APL cell lines derived by x-ray mutagenesis302 were found to lose expression of the PML-RARα protein, although the fusion gene and mRNA remained.302 This was due to a protease activity that degrades exogenous PML-RARα and is blocked by chemical inhibitors of the proteosome.303 This highlights the importance of the PML-RARα protein both in generating the APL phenotype and in mediating the unique sensitivity of the disease to ATRA. These data also imply that there are secondary changes in APL cells that maintain the transformed phenotype even in the absence of stable expression of the protein.

Resistant APL cells were also generated by prolonged culture of NB4 cells121 in the presence of ATRA.304,305 One such resistant cell line expressed the PML-RARα protein but had an abnormal retinoic acid binding profile305 and failed to upregulate tissue glutaminase II expression in response to ATRA. This was due to a missense mutation in the E domain of PML-RARα,306 which abolished its ability to bind ligand and mediate trans-activation by ATRA. These data support the notion that PML-RARα has a critical effect in blocking gene expression at low doses of ATRA and further underscores the fact that therapeutic response to ATRA in APL is dependent on the ability of the chimeric protein to activate transcription in the presence of ligand, reversing the blockade of target genes. In this and other resistant cell lines, at least one gene, CD18, continued to be induced by ATRA. This indicates that the remaining endogenous RARs within the cell can activate a subset of target genes, but the genes most critical for cell differentiation continue to be inhibited by PML-RARα.

These types of mutations are clinically relevant, because recent studies showed mutations in the ligand binding domain or adjacent AF-2 region in nearly 15% of patients both de novo but particularly after prolonged ATRA treatment,307,308 indicating that ATRA treatment puts a strong selective pressure for clones with defects in PML-RARα.308 However, the sequence of PML-RARα in cell lines derived from resistant patients and in primary patient specimens is most frequently normal, indicating that another mechanism must play a major role in ATRA resistance, such as loss of the PML-RARα fusion protein due to accelerated degradation or activation of novel oncogenes.303,308 309 

Whereas PML is localized in 6 to 30 large nuclear bodies/cell measuring between 0.2 and 0.3 μm191 in APL, it is delocalized to greater than 100 small (0.1 μm) microspeckles197 due to the ability of PML to heterodimerize with PML-RARα though the coiled-coil motif.170,191 PML-RARα draws other nuclear proteins, including SP100,198 PLZF,216RXR,197 and Rb,229 into the microspeckled structure as well. These microspeckles have no evident structure198 and colocalize with nascent RNA, signifying the transcriptional function of PML-RARα.191,201 In at least one set of studies, a large proportion of PML-RARα fusion was localized in the cytoplasm rather than the nucleus.173 This is consistent with the notion that PML-RARα, under low physiological concentrations of retinoids, acts as a dominant negative receptor drawing critical factors away from loci controlled by RARα to a new set of loci or to a transcriptionally inactive compartment.

ATRA treatment of APL cells relocalizes the PML protein into the wild-type nuclear body configuration.173,191,197,198,310This is largely due to degradation of PML-RARα95,96,191,311,312 through the action of the proteosome,303 likely by the induction of a caspase 3-like activity after ATRA treatment.313 The specific cleavage of PML-RARα occurs C-terminal to the the RING, B-boxes, and coiled-coil motifs of PML, yielding a product recognized by RARα antibodies that contains residual PML sequences.95,96 The resulting protein could be predicted to be unable to bind wild-type PML, which would then be released and free to form its usual macromolecular complex in the NB. The remaining truncated PML-RARα protein might function in a similar fashion as wild-type RARα, activating its target genes and no longer sequestering other proteins critical for cell differentiation through the N-terminal PML moiety. However, early after ATRA treatment, reactivity to RARα antibodies is detected in the large nuclear bodies, suggesting that the PML-RARα protein itself undergoes some conformational shift or novel protein association after ATRA treatment, which then allows it to colocalize with wild-type PML in the NBs.173,197,198 It should also be noted that the PML(S)-RARα isoform does not contain the sequences required for caspase cleavage and does not undergo degradation after ATRA, yet these patients respond to ATRA therapy.313,314 In addition, in a model system, ATRA could induce differentiation even in the presence of caspase inhibitors, suggesting that the degradation of PML-RARα may not be essential for therapeutic response.313 The relatively slow reorganization of the NB is accelerated by other agents, such as cyclic AMP,310 which also increase the rate of differentiation, suggesting that altered phosphorylation of the PML-RARα fusion changes its rate of degradation and/or relocalization. Thus, PML-RARα relocalization is highly correlated with induction of differentiation in APL cells, suggesting that disruption of some component of the nuclear body other than PML plays a key role in this process.

Other agents promote normalization of the nuclear body structure through different mechanisms. Recent studies indicate that arsenic trioxide (As2O3), a component of traditional Chinese medicine, induces complete clinical remission of APL in ATRA resistant patients.315 Whereas reaggregation of the nuclear body takes 1 to 2 days of ATRA exposure, treatment with As2O3 leads to rapid formation of wild-type pattern nuclear bodies within 6 hours, followed by loss of PML staining after 24 hours.311,316,317 This effect was observed both in NB4 and other cell lines and was enhanced by IFN.179,311During this process, both PML and PML-RARα are targeted to the nuclear body and then rapidly degraded, although there is no effect on other NB proteins such as SP100 and relatively little degradation of endogenous RARα.318,319 Furthermore, As2O3 increased the transfer rate of PML from the nucleoplasm to the nuclear matrix179,311 and increased PML levels within the NB, accelerating apoptosis.252 It was recently shown that As2O3 induces the phosphorylation-dependent covalent linkage of PML to the ubiquitin-like molecule sentrin, perhaps targeting the protein for degradation. The C-terminal portion of PML was required for its targeted degradation. Whether sentrin binds PML-RARα is controversial and whether sentrinization or ubiquination of PML-RARa is required for its degradation is not yet certain.180,227As2O3 induces degradation of PML-RARα even in APL cell lines resistant to ATRA.318-321 Both in vitro and in cells derived from patients undergoing As2O3treatment, this correlates with only partial differentiation of the malignant promyelocytes and predominantly the induction of apoptosis.311,318-320 Simultaneous treatment with ATRA and As2O3 enhanced differentiation and apoptosis of NB4 cells319 and enhanced survival of animals harboring APL (H. de Thé, personal communication, December 1998). However, this was not the case in fresh human APL cells318 treated in vitro, making it not yet certain whether ATRA and arsenic might best be used concommitantly or as sequential agents in the treatment of APL. The organic arsenical melarsoprol may also provide effective treatment of APL321 and other hematological malignancies.322 As2O3 and melarsoprol induce apoptosis of an APL cell line without detectable PML-RARα as well as PML−/− murine fiboblasts.321 However, others have found a dependence of PML expression for arsenic induced apoptosis (H. de Thé, personal communication, December 1998), and one group found that a cell line harboring PLZF-RARα could not be induced to undergo apoptosis with arsenic,227 suggesting that the extreme sensitivity of APL cell lines to arsenic may be due to their dependence on PML-RARα for continued growth and the prevention of apoptosis. Intriguingly, antimony, a metal in the same column of the periodic table as arsenic, can also induce the degradation of PML and induction of apoptosis of APL cells,323 suggesting a common mechanism of covalent modification of critical cellular proteins by these heavy metals.

These results lead to a model in which treatment of APL can occur by two different mechanisms, each rescuing the disrupted nuclear body and perhaps restoring a growth control mechanism to the cell. In both cases, degradation of PML-RARα releases the complete block on cell differentiation. After both ATRA88 and As2O3315 treatment, an initial hyperleukocytosis is noted. However, the complete lack of PML-RARα after As2O3 treatment leads to apoptosis, whereas the residual PML-RARα fragment present after ATRA treatment, in combination with residual RARα, induces the genes critical for cell differentiation. How these agents cause the retargeting of PML-RARα to the nuclear body is unknown. As2O3 can react with sulfhydryl groups and alters phosphorylation pathways, a fact supported by the finding that As2O3 treatment is associated with hyperphosphorylation of the RARα itself.311 ATRA may cause a conformational shift in PML-RARα, allowing new sets of proteins to interact with the ligand binding domain. RXR may play a role in this conformation shift as well. When treated with ATRA plus an RARα antagonist, APL cells did not differentiate or reorganize their NBs. However, when treated with the same RARα antagonist plus a RXR agonist, both NB reorganization and differentiation ocurred.68 These experiments underline the complexity of retinoid signaling and highlight the importance of both components of the RAR/RXR heterodimer in myeloid differentiation and APL pathogenesis.

Although the disruption of the nuclear body in t(15;17) APL is one of the most dramatic features of this disease, it may not be absolutely required for the pathogenesis of APL, because PML is in the wild-type configuration in variant forms of APL. Hence, it may be the degradation of PML-RARα rather than changes in PML/NB function that may be most critical for the induction of differentiation in APL. Alternatively, a component of the NB other than PML may be sequestered by all of the RARα chimeras.

Cellular models of PML-RARα function have been hampered by the toxicity of the fusion protein, as underscored by studies in which the PML-RARα fusion protein could not be expressed after retroviral infection in nonhematopoietic cell lines and was expressed in only a few hematopoietic cell lines. PML-RARα retroviruses were difficult to generate due to the growth-suppressive effects of the protein on fibroblasts, including retroviral packaging cell lines.324-326 

The most successful model of PML-RARα function in APL was constructed in the monocytoid U937 cell line.324 Cells stably or inducibly expressing PML-RARα failed to differentiate in response to ATRA or a combination of vitamin D3 plus transforming growth factor β (TGFβ).324,327 Under physiological concentrations of ATRA (10−9 mol/L), PML-RARα expression was associated with an increase in cell growth rate. However, when treated with 10−6 mol/L ATRA, PML-RARα expression was associated with markedly decreased cell proliferation and increased differentiation. The change in growth rate was due to an increase in apoptosis and not to alterations in the cell cycle. In addition, when grown under conditions of reduced serum, PML-RARα–expressing U937 cells proliferated, whereas control cells underwent apoptosis. PML-RARα also blocked apoptosis in response to TNFα.328 TNF resistance was due to a posttranscriptional downregulation of the TNF receptor, allowing the APL cell to escape an autologous growth inhibitory mechanism, because APL cells secrete high levels of TNFα.329 From these data it was proposed that PML-RARα might function to promote cell survival. This idea was supported by the fact that myeloid, G-CSF–dependent TF-1 cells expressing PML-RARα were protected from apoptosis induced by G-CSF withdrawal.314,330 In addition, recent data indicate that ablation of PML-RARα expression in NB4 APL cells either by homologus recombination or expression of a ribozyme induces apoptosis,331,332 even in ATRA-resistant cells. The fraction of cycling cells in APL is relatively low; thus, the persistence of cells due to the anti-apoptotic effects of PML-RARα could be critical. If PML-RARα prevents apoptosis when cells are grown under physiological conditions of ATRA, how does the protein encourage apoptosis when such cells are treated with phamacological ATRA or As2O3?311,315,318,319,333One possibility is that PML-RARα is degraded under these conditions,96,311 removing the protective agent. Alternatively, these agents, through the action of PML-RARα, may activate novel target genes that play a role in cell cycle arrest and apoptosis. The p21 gene is such a candidate, being activated by both ATRA130 and arsenic.319As2O3-mediated apoptosis of APL cells is accelerated by agents that deplete cellular glutathione, a scavanger of free radicals.334 Given recent data that p53 induces apoptosis through generation of reactive oxygen species,335there may be overlap between the p53 and As2O3apoptotic pathways.

One of the drawbacks of the U937 model is that these cells undergo monocytic rather than granulocytic differentiation. One model, possibly more reflective of the pathophysiological role of PML-RARα, was created by transient expression of PML-RARα in HL60 cells. This inhibited granulocytic differentiation induced by ATRA and vitamin D3 but not granulocytic differentiation induced by dimethyl sulfoxide (DMSO) or monocytic differentiation induced by phorbol ester. These results support a relatively specific mechanism of action for PML-RARα upon nuclear receptor pathways. However, NB4 cells cannot differentiate in response to vitamin D3,300,327 but when ATRA and D3 are added in concert or sequentially (ATRA first), marked differentiation and inhibition of proliferation occurs. In addition, NB4 cells are resistant to polar compounds such as sodium butyrate or HMBA, unless pretreated with ATRA for a period as brief as 30 minutes.300 Thus, PML-RARα may affect non-nuclear receptor differentiation pathways as well (see below). Recently, PML-RARα was shown to enhance the proliferation of murine bone marrow progenitor cells after retroviral transfer, allowing these cells to be serially replated ex vivo. However, the cells remained growth factor dependent, suggesting that PML-RARα on its own cannot completely transform the cell. The cell lines resulting from PML-RARα expression were undifferentiated and could be induced to stop proliferation and undergo differentiation after ATRA treatment.336 

The U937 model was used to determine which structural features of the PML-RARα fusion protein were critical for effects on cell growth and differentiation.163 PML expression did not block differentiation induced by vitamin D3 and TGFβ, whereas RARα did slightly, perhaps due to sequestration of RXR. PML-RARα was a more potent inhibitor of differentiation. Deletion analysis showed that the first coiled-coil motif of PML-RARα was required for its ability to block differentiation, but deletion of this segment did not affect PML/PML-RARα interaction, NB disruption, or RARE-dependent trans-activation. This suggests that disruption of PML within the nuclear body is not critical for the action of PML-RARα and that the ability of PML-RARα to interact with an unidentified factor, through the coiled-coil motif, may be critical for its function. In support of this hypothesis, deletion of coil 2 prevents PML-RARα from delocalizing PML from the NBs, but still allows disruption of the nuclear body pattern of Sp100. This protein or another of the multiple proteins within the nuclear body such as Rb229 could represent the target protein of PML-RARα. This is not likely to be RXR, because coexpression of RXR prevented differentiation block induced by wild-type RARα but not the block mediated by PML-RARα. The RING finger and B box motifs are also always found in the PML-RARα fusion protein of APL patients, suggesting that the integrity of the RBCC unit is required for leukemogenesis. Furthermore, the ability of PML-RARα to induce apoptosis in nonhematopoietic cells depends on the integrity of the RING finger/B-box motifs and the microspeckled localization of PML-RARα. How this relates to APL pathogenesis by PML-RARα is unclear.326 Lastly, as noted, there are subtle differences in the PML(L)-RARα and PML(S)-RARα isoforms. In TF-1 cells, only the long PML isoform inhibited cell growth, whereas only the short isoform protected the cells from growth factor withdrawal. When cells were treated with ATRA, they underwent apoptosis, with the long isoform showing a more prominent effect.314 

Several major conclusions can be reached from these cellular models. (1) PML-RARα does not activate cell growth or confer factor-independent growth to cells and in this regard is not a conventional oncogene. (2) PML-RARα alters the cell setpoint for apoptosis. It inhibits apoptosis due to growth factor withdrawal in hematopoietic cells, but encourages it in the presence of ATRA or arsenic. In many nonhematopoietic cells, PML-RARα is toxic and induces apoptosis. (3) PML-RARα can inhibit differentiation mediated by nuclear receptor pathways and some non-nuclear receptor pathways. The mechanism of cross-talk with nonreceptor pathways is unknown. (4) PML-RARα requires the first coiled domain of PML and the AF2 domain, which binds HDAC complexes, of RARα to block differentiation. The second coiled domain, which delocalizes PML, is dispensible for differentiation block. The first coiled-coil motif of PML may contact a critical cofactor. (5) The PML(S)-RAR isoform, which lacks phosphorylation sites, and the nuclear localization sequence of PML may have somewhat different biological properties from the PML(L)-RARα isoform.

Several laboratories have tried to model APL in animals. Infection of avian bone marrow with a PML-RARα–containing retrovirus resulted in an undifferentiated form of leukemia that did not respond to ATRA.337 Curiously, the PML-RARα gene harbored in these leukemic clones bore two point mutations. The first, located in the RING finger, caused the fusion protein to lose the typical microspeckled pattern and have altered transcriptional properties. The second replaced a serine with an alanine residue in the coiled-coil domain, causing the loss of a potential phosphorylation site. These studies, although successful in demonstrating the oncogenic potential of PML-RARα, highlight the difficulties of working with this toxic protein.326 Interestingly, infection of mouse bone marrow with a C-terminal truncated form of RARα also yields the outgrowth of a lymphohematopoietic precursor cell, indicating that there may be two points of control by ATRA in hematopoiesis, one at the pluripotent progenitor stage and a second at the promyelocyte stage.338Why RARα disruption clinically yields only the promyelocyte phenotype is uncertain.

The first transgenic mouse created that expressed the PML-RARα fusion used the CD11b promoter, which was expressed relatively late in myeloid maturation. These mice did not develop APL or a preleukemic syndrome, but did show a defect in myeloid precursor response to cytokines and profound neutropenia after sublethal irradiation, implying that the PML-RARα protein impaired myeloid development.339 Another transgenic murine model expressed the PML-RARα transgene from the metallothionine promoter.340 The investigators had difficulty obtaining transgenic founders, likely reflecting the poor tolerance of PML-RARα by nonhematopoietic cells. One animal line expressed PML-RARα only in the liver and brain, which was induced by the addition of Zn2+ to the animals’ drinking water. These animals developed liver pathology, including hepatocellular carcinoma after only 5 days of induction with zinc. ATRA treatment did not prevent the emergence of liver disease, which was associated with an increased proliferation rate and no change in spontaneous apoptosis. These experiments confirmed the oncogenic nature of PML-RARα and suggested that, under certain circumstances, the protein could accelerate cell proliferation. Two groups performed transgenic experiments with PML-RARα using the cathepsin G promoter.341,342 These mice developed a preleukemic syndrome characterized by an increase in immature myeloid forms in the bone marrow and splenomegaly due to extramedullary hematopoiesis.341 About 10% to 30% of the animals developed leukemia, with a median latency of 300 days; it was also associated with anemia, thrombocytopenia, and massive splenomegaly. A modest peripheral promyelocyte count of only 7% was noted,342 with some cells harboring Auer rods. No bleeding diathesis was noted. Unlike human APL, many fully differentiated granulocytes were present in the peripheral blood before treatment with ATRA. ATRA treatment led to an initial increase in peripheral white blood cell count, reminiscent of the retinoic-acid syndrome88 and consistent with the mobilization of cells from the marrow. This was followed by a decrease in the leukocyte count and the appearance of differentiated neutrophils. However, after ATRA treatment, Grisolano et al341 found that promyelocyte counts decreased and the cells appeared to undergo apoptosis rather than differentiation. Therefore, whether ATRA induced differentiation of the promyelocytes or selectively killed the immature cells is not certain, because differentiated cells were present both before and after ATRA treatment. These animals therefore offer a somewhat imperfect model for differentiation therapy. Intriguingly, as in humans,7,8 all mice relapsed even with the continuation of ATRA, suggesting that other oncogenic lesions must be involved in APL development.299 

The mouse model of APL most similar to human disease was generated by use of the MRP8 promoter, which is expressed at the promyelocyte to metamyelocyte stage and continues to be active in mature neutrophils,343 as opposed to the cathepsin G gene, which is expressed during a more narrow window of promyelocyte differentiation.344 These mice also developed a preleukemic phase, and about one third developed promyelocytic leukemia with a median latency of 6 months345 (S. Kogan, personal communication, July 1998), accompanied by bleeding, anemia, thrombocytopenia, and a low leukocyte count, all characteristic of human APL. When these cells were placed into culture and treated with ATRA, differentiated neutrophils were observed. The transgenic mice also developed epidermal papillomas, which further demonstrated the neoplastic activity of the PML-RARα fusion. When the animals were treated with ATRA, mature neutrophils appeared in the peripheral blood and marrow and splenomegaly was reduced, consistent with clinical differentiation. Highly purified, residual nonleukemic progenitor cells from APL patients are PML-RARα negative,346suggesting that the PML promoter driving PML-RARα expression must function at a specific stage of myeloid differentiation. Therefore, a knock-in347 strategy for the creation of transgenic mice might yield the most physiologic model of t(15;17) translocation APL.

In all of these models, the delay in onset of leukemia suggests that a second, as yet uncharacterized, genetic hit is required for neoplastic transformation. Whether the leukemias that do develop in the mice are monoclonal or polyclonal has not yet been determined. A monoclonal origin would be consistent with a model of tumor promotion induced by PML-RARα, potentially by preventing apoptosis and blocking differentiation, followed by a second genetic lesion. Such a multistep pathway was modeled by the introduction of N-ras into the hematopoietic cells derived from mice harboring the PML-RARα fusion expressed from the CD11b promoter.348 As a result, there was a 10- to 100-fold synergistic increase in myeloid colonies. With this proof of principle, we can anticipate experiments in which PML-RARα mice could be crossed with mice expressing activated oncogenes or lacking tumor suppressors. The APL that develops spontaneously in PML-RARα mice could also be screened for secondary mutations required for pathogenesis.

Myeloid differentiation usually occurs at physiological levels of ATRA (10−8 mol/L), which activates key RARα target genes. These target genes may contain high-affinity RAREs or bind factors that cooperate with the RARα to load the basal transcriptional machinery onto the promoter, even when only a fraction of cellular RARα is engaged by ligand. The highly expressed PML-RARα protein may functionally sequester RARα cofactors from the wild-type receptor or bind to critical genes in place of RARα. Even if a PML-RARα/RXR heterodimer bound to the critical genes, the increased affinity of PML-RARα for corepressors294 299 would make it a poor activator of RARα target genes. This would overwhelm the usual tonic effect of RARα to induce myeloid differentiation at low ATRA concentrations (Fig 5).

Fig. 5.

Suggested model of PML-RAR action in APL. (A) At 10−9 to 10−8 mol/L ATRA, PML-RAR prevents activation of key target genes required for myeloid differentiation by sequestration of RXR and other RAR cofactors, inhibiting normal RAR function. In addition, PML-RAR may bind to RAR targets as a homodimer or as a heterodimer with RXR and inhibit transcription of these genes by recruitment of corepressor/histone deacetlyase complexes. PML-RAR also may affect transcription mediated by AP1 and IFN-responsive factors and can sequester PLZF and potentially affect its function. PML-RAR prevents apoptosis through unknown mechanisms and delocalizes PML and other proteins from the nuclear body, although the importance of this is uncertain, because the NB is normal in the other forms of APL. (B) In the presence of pharmacological doses of ATRA, the PML-RAR fusion is degraded and releases PML and other cofactors. The NB structure is restored. A residual fragment of the PML-RAR fusion and/or the wild-type RAR, which is upregulated in response to ATRA, can then stimulate transcription of myeloid target genes. The blockade of other signaling pathways is released and the anti-apoptotic effect of PML-RAR is lost. As a result, terminal cell differentiation can proceed.

Fig. 5.

Suggested model of PML-RAR action in APL. (A) At 10−9 to 10−8 mol/L ATRA, PML-RAR prevents activation of key target genes required for myeloid differentiation by sequestration of RXR and other RAR cofactors, inhibiting normal RAR function. In addition, PML-RAR may bind to RAR targets as a homodimer or as a heterodimer with RXR and inhibit transcription of these genes by recruitment of corepressor/histone deacetlyase complexes. PML-RAR also may affect transcription mediated by AP1 and IFN-responsive factors and can sequester PLZF and potentially affect its function. PML-RAR prevents apoptosis through unknown mechanisms and delocalizes PML and other proteins from the nuclear body, although the importance of this is uncertain, because the NB is normal in the other forms of APL. (B) In the presence of pharmacological doses of ATRA, the PML-RAR fusion is degraded and releases PML and other cofactors. The NB structure is restored. A residual fragment of the PML-RAR fusion and/or the wild-type RAR, which is upregulated in response to ATRA, can then stimulate transcription of myeloid target genes. The blockade of other signaling pathways is released and the anti-apoptotic effect of PML-RAR is lost. As a result, terminal cell differentiation can proceed.

Close modal

In the presence of pharmacological doses of ATRA, the PML-RARα fusion releases the corepressors and stimulates transcription of target genes that allow myeloid development to proceed. Furthermore, the PML-RARα protein is degraded95,96 and wild-type RARα is upregulated,94 shifting the balance of power of RARs in the cell from PML-RARα to RARα. However, the fact that PML-RARα can confer ATRA sensitivity to mutant HL60 cell lines without endogenous wild-type RARα suggests that PML-RARα does mediate at least part of the pro-differentiative349 effect. A PML-RARα/RXR heterodimer is probably the mediator of differentiation, because this process is synergistically stimulated by a combination of RARα- and RXR-specific ligands.68,76 In addition, brief treatment of NB4 cells or fresh APL cells with ATRA allows potent differentiation of APL cells to proceed in the presence of other agents such as hexamethylene bis acetamide (HMBA), cyclic AMP, and vitamin D3,300 350 suggesting that rapid transcriptional events before PML-RARα degradation mediate differentiation. These events might also include activation of other nuclear receptors, STATs and AP1. In contrast, arsenic degrades PML-RARα fusion without RARα-mediated signaling. Modest differentiation might occur in this case by low level signaling through the endogenous RARα. A component in the pathogenesis of APL may include the delocalization of one or more key proteins from the NB. Current evidence points away from this being PML itself.

The reciprocal RARα-PML fusion generated in t(15;17)11,351 is present in 70% to 80% of APL cases351,352 (Fig 3). Two transcripts can be generated from the alternative RARα promoters, of which RARα1-PML was the most common.353 As in the case of PML-RARα, several different forms of RARα-PML were also found due to alternative breakpoints within the PML gene. RARα1-PML and RARα2-PML transcripts from patients with breakpoints 5′ in the PML gene encode RARα-PML proteins, whereas fusion transcripts derived from a more 3′ breakpoint in the PML gene only encode a truncated peptide containing a portion of the RAR A domain. RARα-PML contains the A1 or A2 domain of the RARα protein fused to a variable portion of the PML protein, due to alternative splicing, including the serine-rich C-terminal domain. It is hard to predict the effects of RARα-PML, because the role of the C-terminus of PML is unknown. There were cases of APL associated with interstitial, nonreciprocal fusions of the PML and RARα genes that did not generate an RARα-PML fusion gene.262-264Furthermore, there is no difference in ATRA sensitivity or clinical outcomes of patients who do or do not harbor the RARα-PML transcript.352,353 Patients with prolonged remissions of APL may express the RARα-PML transcript and not the PML-RARα transcript, suggesting that the small number of cells that harbor these genes may not have leukemogenic potential,354 although a sole APL patient was reported to posses the RARα-PML and not the PML-RARα transcript.355 In general, RARα-PML does not appear to be required for the pathogenesis of APL. Although transgenic mice harboring the RARα-PML fusion did not develop leukemia, when crossed with PML-RARα mice, leukemia developed with greater frequency.356 Hence, the RARα-PML may contribute to the disease process.

The PLZF gene.

The PLZF gene was initially identified by its rearrangement in an APL patient from Shanghai with translocation (11;17)(q23;q21)357-359(Fig 6). Eight confirmed cases of t(11;17)(q23;q21) APL fusing the PLZF and RARα genes were described,360-363 and a recent workshop identified a total of 8 cases around the world.364 Morphologic review of the original 6 patients and the 8 cases of the workshop showed features intermediate between M2 and M3 leukemia, with sparser granules, a lack of Faggot cells, and the absence of a bilobed nucleus.361Strikingly, these 6 patients were resistant to differentiation therapy with ATRA as well as chemotherapy. Leukemic cells from these patients could not be induced to differentiate with ATRA in vitro.360,361 Translocation (11;17)(q23;q21)-APL is unique in its resistance to ATRA, suggesting a critical difference between PLZF and the other four fusion partners of RARα in APL (Table 4). However, in opposition to this view, cells from 1 patient with t(11;17)(q23;q21) APL could be induced to differentiate when treated with a combination of ATRA and G-CSF but not ATRA alone,365 and this patient was successfully treated with a combination of ATRA and G-CSF.366 One other patient achieved a remission with concurrent ATRA and chemotherapy,363 suggesting that the resistance of this form of APL may not be absolute.

Fig. 6.

Functional domains of the PLZF protein and structure of the PLZF-RAR and reciprocal RAR-PLZF proteins generated in t(11;17)(q23;q21) APL. PLZF-RAR always contains the N-terminal POZ/BTB-self-association domain. Heterogeneity in the PLZF gene breakpoint can yield PLZF-RAR fusion proteins, including either the first two or first three zinc fingers of PLZF. The reciprocal RAR-PLZF protein can bind to PLZF binding sites and contains the last six or seven of the nine PLZF zinc fingers linked to the AF1 activation domain of RAR rather than the PLZF repression domains.

Fig. 6.

Functional domains of the PLZF protein and structure of the PLZF-RAR and reciprocal RAR-PLZF proteins generated in t(11;17)(q23;q21) APL. PLZF-RAR always contains the N-terminal POZ/BTB-self-association domain. Heterogeneity in the PLZF gene breakpoint can yield PLZF-RAR fusion proteins, including either the first two or first three zinc fingers of PLZF. The reciprocal RAR-PLZF protein can bind to PLZF binding sites and contains the last six or seven of the nine PLZF zinc fingers linked to the AF1 activation domain of RAR rather than the PLZF repression domains.

Close modal
Fig. 7.

Crystal structure at 1.9Å of the PLZF BTB/POZ domain dimer382 as viewed from the side.

Fig. 7.

Crystal structure at 1.9Å of the PLZF BTB/POZ domain dimer382 as viewed from the side.

Close modal

The PLZF gene, localized on chromosome 11q23, yields a 7-kb mRNA that codes for a zinc finger transcription factor.289,359,367,368 The PLZF gene is 1 Mb telomeric to the MLL/HRX gene, which is frequently rearranged in leukemia.369-371 The genomic structure of PLZF is incompletely known, but it is clear that the N-terminal portion is encoded on a single exon and that the C-terminal zinc finger motifs are encoded by a number of small exons (Baysal et al372 and Z. Chen, personal communication, June 1998). PLZF codes for a DNA-binding protein of 673 amino acids with nine Krüppel-like C2H2 zinc finger domains with a predicted molecular weight of 74 kD (Fig 6) that migrates through polyacrylamide gels with an apparent MW of 80 to 90 kD.

The N-terminal 118 amino acids encode a POZ (Pox virus and Zinc finger) or BTB (Broad Complex, tramtrack, Bric a Brac) domain. The POZ/BTB domain mediates protein self-association and heterotypic associations373 and acts as a transcriptional repression domain within zinc finger374-377 and basic-zipper378 transcription factors. The POZ/BTB domain can be involved in chromatin remodeling and histone mobilization379,380 and transcriptional repression through interaction with histone deacteylase (see below). Physical characterization of the PLZF POZ/BTB domain found it to be a tight dimer in solution (Kd = 2 × 10−7), with a high amount of helical structure.381 Characterization of a 1.9Å crystal structure of the BTB/POZ domain of PLZF382confirmed these findings, showing a highly intertwined dimer with a large hydrophobic interface. The top portion of the dimer structure forms a groove exposed to solvent lined with conserved charged amino acids potentially representing a peptide binding site (Fig 7). Several missense mutations of conserved residues interfere with dimer contacts and disrupt the ability of the POZ domain to repress transcription, suggesting that dimerization and repression may be closely linked.605 

In transfected cells, PLZF is localized to the nucleus and is289,368 phosphorylated on serine and threonine residues (Shaknovich and Licht, unpublished data). MDS, a primitive leukemic cell line, expresses high levels of PLZF mRNA and PLZF protein when treated with the calcium ionophore A23187.289Confocal microscopy of MDS cells showed that PLZF localized to approximately 50 small nuclear subdomains, with a lower level of diffuse nuclear staining also noted, whereas only 10 PML-containing NBs were noted per cell. The speckled pattern of subnuclear expression of PLZF depends on the presence of the POZ/BTB domain as removal of this domain leads to expression in a diffuse nuclear pattern.383Given the potential importance of the expression of the PML protein in the NB, it became important to determine whether PLZF was expressed in the same or similar domains. In hematopoietic cells that naturally express both PML and PLZF as well as in transiently transfected nonhematopoietic cells, wild-type PML and PLZF could colocalize in nuclear body structures. However, this colocalization was not complete, indicating that PLZF may act in both the nuclear body and other subnuclear compartments.216 The PLZF-RARα fusion protein, either transfected into cells or naturally expressed in blasts from a patient with t(11;17)(q23;q21) APL, did not colocalize with PML or delocalize PML from nuclear body structures.216 This critical fact indicates that delocalization of the PML component of the nuclear body is not required for the pathogenesis of APL. In contrast, in t(15;17) APL cells, PLZF is delocalized into a microspeckled pattern identical to PML-RARα.216,299 The coiled-coil region of PML, which is also responsible for PML self-association, was required for the PML-PLZF interaction. The first two zinc fingers of PLZF and not the POZ/BTB domain mediates the interaction of PLZF with PML (A. Zelent, personal communication, December 1998). Therefore, PML and other related RING finger proteins might be cofactors for PLZF function. Delocalization of PLZF might be a common and central theme in the pathogenesis of APL, because PLZF is also found in an abnormal microspeckled pattern and in t(5;17)-APL associated with the NPM-RARα protein.384 

PLZF mRNA is expressed in myeloid cell lines such as KG1, HL60, and NB4 as well in as the multipotent cell line FDCPMixA4. It is expressed at lower levels in more differentiated erythroleukemia, promyelocytic, and monocytic cell lines359,368 as well as in peripheral blood mononuclear cells,359 tissue macrophages (Fallon and Licht, unpublished data), and pro-B–cell lines.368PLZF is also expressed in mature B-cell and chronic lymphocytic leukemia (CLL) specimens.385 PLZF is downregulated during ATRA-mediated differentiation of NB4 and HL60 cells (Chen et al359 and Chen et al, unpublished data) and during differentiation of FDCPMixA4. In contrast, PLZF is upregulated in the MDS cell line after treatment with calcium ionophore, perhaps recapitulating some aspect of monocyte development.289 In embryonic stem cells, PLZF levels increase as the cells are allowed to form embryoid bodies that presumably contain hematopoietic elements. Finally, CD34+human progenitor cells could be immunostained with PLZF antisera in a distinct nuclear speckled pattern.368 When such cells were placed into culture and allowed to differentiate, PLZF levels transiently increased then declined (C. Labbaye, personal communication, December 1998). Incubation of human bone marrow with antisense PLZF oligonucleotides led to a decrease in the number of burst-forming unit-erythroid (BFU-E) and colony-forming unit–granulocyte-macrophage (CFU-GM) colonies (Shaknovich et al, unpublished data). Taken together, these data indicate that PLZF expression may be important for the maintenance or survival of hematopoietic stem cells and or early progenitors. Scheduled, regulated downregulation of PLZF may be required for normal hematopoietic differentiation and proliferation.

In the murine embryo, PLZF is expressed in the aorta, gonadal, mesonephros region (AGM), a zone containing hematopoietic precursors. Also, during mouse embryogenesis, PLZF is prominently expressed in the developing neural tube. Expression throughout the central nervous system (CNS) is initially uniform at day 8.5 postconception (pc). Subsequently expression is downregulated in rhombomeric segments 3 and 5 at the same time that genes such askrox-20 and hoxb2 are upregulated in these segments.367 A PLZF site was found in the hoxb25′ flanking region and PLZF could repress the hoxb2promoter in cotransfection assays,386 suggesting that PLZF might directly regulate this gene. By 10 days pc, PLZF expression is restricted to the boundaries of the rhombomeres, perhaps acting to limit the expression of critical pattern formation genes. Early widespread expression of PLZF may initially repress developmental programs within the CNS, and the selective downregulation of PLZF could lead to segmental identities.387 Similarly, PLZF may repress the differentiated phenotype of myeloid cells, and its downregulation may allow differentiation to proceed (see below). It is reasonable to theorize that altered regulation of homeobox genes may be central to PLZF action in the development of both the CNS and hematopoietic system. Other sites of PLZF expression include neural crest cells, branchial arches, facial processes, and apical epidermal ridges of the developing mouse and chick limb buds367,387(C. Tabin, personal communication, July 1998). The latter are sites of signaling between the epithelium and the underlying mesenchyme and suggest a role for PLZF in limb patterning. PLZF is also expressed in the mesonephros, a precursor kidney structure, and in the dilated structures found in autosomal dominant polycystic kidney disease (P. Wilson and J. Licht, unpublished data), which are felt to partially recapitulate early renal development. Murine PLZF expression is considerably upregulated in the liver, heart, and kidney in the perinatal period and shortly after birth. In mutant albino mice that have a defect in tyrosine metabolism PLZF, along with a number of liver-specific factors, including HNF-1, HNF-4, and C/EBPα, fail to be induced at birth, grouping these genes in a common regulatory pathway.367 Lastly, a search of the EST database (http://www.tigr.org) indicates that PLZF is expressed in both skeletal muscle and adipose tissue. In summary, although isolated by its involvement in APL, PLZF may play a role in nervous system development and limb patterning, renal development, hematopoietic development, and energy metabolism. Preliminary analysis of PLZF knockout animals388 found that they weighed up to 40% less than heterozygous littermates and have abnormally kinked tails and multiple skeletal defects, including foreshortening of the limb and fused digits. This suggests that PLZF, like RARα, might affect the expression of Hox genes involved in limb and body patterning. Additionally, PLZF may influence genes responsible for apoptosis limb and digit development. Homozygous PLZF null mice are sterile with testicular hypotrophy and impaired spermatogenesis. Disruption of other transcription factors, including CREM, RXRβ,389 and A-myb,390 has a similar phenotype, suggesting similar or overlapping functions. Intriguingly, cyclin A, the expression of which is affected by PLZF (see below), is expressed in a distinct developmental pattern in the developing testis.391 392 This suggests that disruption of PLZF expression might affect cell cycle regulators critical for spermatogenesis. PLZF null mice are viable, and, to date, these mice have not exhibited an obvious hematopoietic phenotype and neither have they developed leukemia or other tumors. This does not rule out a role for PLZF in hematopoiesis and could indicate the presence of redundant genes to partially compensate for the lack of PLZF during development.

PLZF is a sequence-specific DNA binding protein that can recognize a TA-rich sequence derived from a pool of randomized oligonucleotides.393,394 A binding site for PLZF was fortuitously discovered in a yeast two hybrid screening experiment. PLZF fused to an acidic activation domain was isolated by its ability to activate a bacterial lex operator-containing reporter gene in yeast.395 The lex operator sequence actually has some similarity to the artificially selected PLZF site, a PLZF binding site within the cyclin A promoter, and a human genomic DNA fragment. With alignment of these sites, a relatively loose consensus sequence of GT (A/C)(A/C) AGT can be derived. The PLZF binding site derived from site selection can be recognized by the C-terminal seven zinc fingers retained in the RARα-PLZF fusion protein. Similarly, the lex operator site can be bound by proteins containing the last 7 or 5 or 4 zinc fingers of PLZF.386,395 The exact role of the first two zinc fingers, which are retained in PLZF-RARα in DNA binding, is not yet clear; they may play a role in associations with proteins such as PML. Given the relatively small length of the PLZF binding site, it is likely that the last 4 finger motifs may directly bind DNA, whereas others may either play a supporting role by interacting with the phosophate backbone in a manner similar to the Gli zinc finger protein,396 whereas others might participate in protein interactions.

Reporter genes containing either the artificial PLZF binding site394 or the lex operator294,397 are repressed by coexpression of the PLZF protein. In contrast, the RARα-PLZF fusion protein can activate a reporter containing a TA-rich PLZF binding site, whereas PLZF-RARα has no effect on these genes. This information suggests that the RARα-PLZF protein could act in a dominant negative manner, binding and altering transcription of PLZF target genes. By fusing portions of PLZF to the heterologous GAL4 DNA binding domain, the PLZF protein was found to contain two separable transcriptional repression domains, one of which overlaps the POZ/BTB domain.394 Similarly, the POZ/BTB domains of the Bcl-6 and ZF5 proteins were found to mediate transcriptional repression.374-377 It is not certain whether the same portions of the POZ/BTB motif are required for PLZF repression, self-association, localization into subnuclear speckles,289,383 and association with cofactors. However, we recently created missense mutations in the POZ/BTB domain that abrogated repression and that will aid in the molecular characterization of the POZ/BTB domain.605 The mechanism of transcriptional repression by PLZF is rapidly becoming elucidated. PLZF was found to interact both in vitro and in vivo with the corepressors N-CoR and SMRT and Sin3A and HDAC1 (histone deacetylase 1).293-295,299,397,398 These interactions occur via the POZ/BTB domain of PLZF, although other regions of PLZF, including the zinc finger motifs, may contibute to binding.296,397,398This correlates with the fact that there are two repression domains in PLZF, with one clearly mapping outside of the POZ domain.394 PLZF, in turn, binds to specific regions of NCoR and SMRT and sin3a and sin3b.295,296,397 SMRT colocalizes with PLZF in nuclear speckles and is able to potentiate the ability of a GAL4-PLZF POZ/BTB domain fusion protein to repress transcription.295 Transcriptional repression by PLZF was potentiated by coexpression of the corepressors and only partially blocked by the HDAC inhibitor trichostatin A,298 suggesting additional mechanisms of repression other than alteration of chromatin.399 Other investigators showed that the Bcl-6 POZ/BTB domain also associates with SMRT400 and other members of the HDAC complex.397 Thus, it could be surmised that many POZ/BTB repressors work similarly by interacting with a multiprotein repressor complex that contains N-CoR, SMRT, sin3A/b, and histone deacetylases, leading to alterations of chromatin configuration.46 Additional mechanisms could be at play as well, because our group found that PLZF forms a DNA-protein complex with a molecular weight of nearly 600 kD that contained cdc2,401 which was implicated in transcriptional repression by phosphorylation of basal transcription factors.402 

PLZF is similar to PML167,184,210,247,248 in that both proteins can repress cell growth. Pools of the interleukin-3 (IL-3)–dependent, nontumorigenic murine myeloid 32DCl3 cells overexpressing the PLZF protein were highly growth inhibited when cultured in IL-3, with their doubling time increasing from 18 hours to greater than 3 days. These cells were retarded in the G1 phase of the cell cycle and had a twofold to threefold increase in the spontaneous rate of apoptosis when grown in IL-3.403 Curiously, PLZF-expressing cells also secreted a negative growth factor into condition cell media that inhibited the growth of non–PLZF-expressing cells. PLZF expression was also associated with inhibition of myeloid differentiation induced by G-CSF or GM-CSF, upregulation of the early hematopoietic marker Sca1, and downregulation of the differentiated granulocytic marker Gr1. The molecular mechanism of action of PLZF on cell growth is beginning to be elucidated. Acute infection of myeloid cells with a PLZF-containing retrovirus was associated with growth arrest of cells in the S-phase of the cell cycle. Progress of cells from G1 into S phase is largely controlled by phosphorylation events mediated by cyclin A paired with CDK2.404,405 Fibroblasts expressing PLZF were growth suppressed and showed blunted induction of cyclin A when stimulated from G0 into the cell cycle by serum. PLZF can bind two sites derived from the cyclin A promoter and can downregulate the cyclin A2 promoter in cotransfection experiments.406 Furthermore, PLZF-expressing, growth-suppressed 32DCl3 cells regain a normal rate of cellular growth when superinfected with a cyclin A-containing retrovirus. These data suggest that cyclin A is a bonafide target gene of PLZF and that PLZF can inhibit cellular growth in a variety of cell types by altering the expression of regulators of the cell cycle.

PLZF expression was associated with protection of 32D cells403 from apoptosis associated with IL-3 withdrawal, suggesting that PLZF might reset the set point between cell life and death, possibly by affecting the expression of bcl-2, bcl-x, bad, or other regulators of apoptosis.407 It is interesting to speculate that high-level PLZF expression plays a role in the quiescence and resistance to apoptosis exhibited by hematopoietic stem cells. Downregulation of PLZF during myeloid differentiation may be accompanied by cycles of committed cell division. It might be argued that PLZF is actually a tumor suppressor disrupted in t(11;17)(q23;q21)-APL. The resulting PLZF-RARα fusion proteins may then act as a dominant negative inhibitors of normal PLZF function (see below). Hence, t(11;17)(q23;q21) APL cells might be functionally null for PLZF. Disruption of PLZF function may also play a role in t(15;17) APL, because PML-RARα delocalizes PLZF in these cells and may thus interfere in its function.216 299 

The t(11;17)(q23;q21) fusion yields two reciprocal transcripts (PLZF-RARα and RARα/PLZF)361,362 (Fig 6). The breakpoint within the PLZF gene occurs 3′ to the first translated exon. As a result of the fusion, the PLZF-RARα chimera contains the entire N-terminal transcriptional effector region of PLZF (including the POZ/BTB domain) as well as the first two zinc fingers of the protein. As in all forms of APL, the RARα gene is fused in the region corresponding to the B domain. In one case, a fusion of the N-terminus of PLZF and the first three PLZF zinc fingers (up to amino acid 484 of PLZF) was linked to RARα, indicating a breakpoint in the PLZF gene further 3′ within the gene. In 4 of 7 cases tested, a reciprocal RARα-PLZF transcript was detected, linking the A/AF1 ligand-independent transcriptional activation domain of RARα42 to the last 7 zinc fingers of PLZF.

In comparing PML-RARα and PLZF-RARα, some interesting contrasts and similarities can be defined. (1) Both fusion proteins can bind as homodimers to RAREs.171,289,383 In the case of PML-RARα, this is mediated by the coiled-coil motif,171 whereas in PLZF the POZ/BTB domain mediates self-association.383 When coincubated with RXR, both PML-RARα and PLZF-RARα form multiple different DNA-protein complexes. It is noteworthy that PLZF-RARα homodimers bound to a direct repeat of the sequence GGG TCA separated by 5 bp (DR5G) with equal avidity as PML-RARα but bound more strongly than PML-RARα to a repeat of the sequence GGT TCA (Dr5T).383 This could potentially explain some of the biological differences between PML-RARα– and PLZF-RARα–associated APL. Although it is possible that PLZF-RARα homodimers might display altered target gene specificity, in the presence of RXR, the PLZF-RARα/RXR heterodimer binds to RAREs in vitro with higher affinity than PLZF-RARα homodimers.289 However, PLZF-RARα, produced by in vitro translation, bound less efficiently to RAREs as a heteromer with RXR than the wild-type RARα.289 This may be due to POZ/BTB domain-mediated multimerization, which could preclude efficient DNA binding (as seen in other POZ/BTB proteins373), suggesting that PLZF-RARα could form complexes that do not efficiently bind to DNA and that could sequester limiting amounts of RXRα, an essential cofactor for RARα function.

(2) Both PML-RARα13,15,157 and PLZF-RARα289,383,408 can act in a dominant negative manner to inhibit the activity of wild-type RARα and the vitamin D3 receptor (Perez et al171 and Licht and English, unpublished data). PLZF-RARα is a relatively weak trans-activator, in some studies completely unable to activate transcription of coexpressed reporter genes408 and in our studies mediating ligand-dependent transcription, albeit at levels less robust than the wild-type RARα.289 The weakened transcriptional activity of PLZF-RARα might be due to inefficient binding by the protein. Alternatively, inclusion of the large PLZF moiety to the N-terminus of the RARα might produce steric hindrance between RARα and coactivators or basal factors. Deletion of the ligand-independent/AF1 activation domain of the RARα reduces activation by RARα, a promoter-dependent effect.42 Most importantly, PLZF-RARα interacts aberrantly with the SMRT and NCoR corepressors, Sin3A and HDAC1, both in vitro and in vivo.293-296,299,397 A key finding relates to the differential affinity of PLZF-RARα and PML-RARα for NCoR and SMRT in the presence of ATRA. Whereas PML-RARα was able to release the corepressors and HDAC1 in the presence of 10−6 mol/L ATRA, PLZF-RARα retained corepressors and HDAC1 even under these high ligand concentrations294-296,299,409(Fig 8). PML-RARα association to HDAC1 and corepressors is mediated solely by the corepressor binding domain or CoR box of the RARα moiety. In contrast, PLZF-RARα binds to corepressors via the CoR box binds and the POZ/BTB domain. It is the latter association that is insensitive to ATRA even at high doses.295,296 This model is supported by several studies. (1) Mutation of the CoR box of PML-RARα but not PLZF-RARα results in loss of binding to HDAC1 and corepressors. (2) Mutation of the PML-RARα CoR box abolishes its ability of this protein to block differentiation, whereas the PLZF-RARα CoR box mutant inhibits differentiation, even in the presence of ATRA.296 (3) HDAC inhibitors, such as trichostatin A and sodium butyrate, were able to convert PLZF-RARα into an ATRA-responsive transcription factor, presumably by inactivating the remaining corepressor complex bound to the POZ/BTB domain. These inhibitors also allowed U937 cells transfected with PLZF-RARα to differentiate in the presence of ATRA.295 296 

Fig. 8.

Potential mechanism of relative resistance of PLZF-RAR–associated APL to ATRA.

Fig. 8.

Potential mechanism of relative resistance of PLZF-RAR–associated APL to ATRA.

Close modal
Fig. 9.

Functional domains of the NPM protein and structure of the NPM-RAR and reciprocal RAR-NPM proteins generated in t(5;17)-APL. A relatively short N-terminal portion of NPM containing the oligomerization domain of the protein is linked to RAR. In the index patient, a longer form of the fusion protein was identified, containing an additional sequences of uncertain origin. The reciprocal RAR-NPM protein could potentially interact with wild-type NPM and interfere with NPM functions.

Fig. 9.

Functional domains of the NPM protein and structure of the NPM-RAR and reciprocal RAR-NPM proteins generated in t(5;17)-APL. A relatively short N-terminal portion of NPM containing the oligomerization domain of the protein is linked to RAR. In the index patient, a longer form of the fusion protein was identified, containing an additional sequences of uncertain origin. The reciprocal RAR-NPM protein could potentially interact with wild-type NPM and interfere with NPM functions.

Close modal

When coexpressed with wild-type RARα, PLZF-RARα inhibits reporter gene trans-activation by the wild-type receptor.289,383,408This dominant negative effect of PLZF-RARα was partially relieved by overexpression of RXRα, consistent with the notion that the aberrant receptors block myeloid differentiation at least partly by limiting the ability of RARα to bind with RXR to its targets.289Deletion mapping of PLZF-RARα protein also showed that dominant negative activity was dependent on the presence of the POZ/BTB domain.383 This region is also required for self-association of PLZF-RARα and for formation of multimers that could sequester RXR. Curiously, inhibition of wild-type RARα function was partially dependent on the presence of the first two PLZF zinc fingers, which are present in the fusion protein and which are also the binding site for PML. When the POZ/BTB domain and first two PLZF zinc fingers were deleted from the fusion protein, PLZF-RARα became an efficient activator of ATRA-mediated transcription.

The dominant negative effect of PLZF-RARα also suggests that it may work by sequestering RARα transcriptional coactivators such as TIF1 or CBP in an inactive conformation, drawing them from RARα target genes. This provides another explanation for why RXR only partially rescues the dominant negative effect of PLZF-RARα. However, the hypothesis that PLZF-RARα binds to RARα corepressors,45,410,411 inappropriately repressing RARα target genes in the absence of ligand is more likely. In keeping with this, PLZF-RARα, like PML-RARα, inhibited the activity of an RARE-containing promoter in the absence of exogenous ATRA.299 Finally, PLZF-RARα could, in theory, also affect the function of wild-type PLZF. In fact, PLZF-RARα and PLZF can preferentially heterodimerize over the formation of PLZF homomeric complexes.383 Hence, high-level expression of PLZF-RARα in t(11;17)(q23;q21) blasts might sequester PLZF from binding to its natural target genes and/or bind to limiting quantities of PLZF transcriptional cofactors.349 However, we have not yet observed this effect in transfection experiments (Li et al, unpublished data).

Cellular models are being developed to clarify the role of PLZF-RARα in APL. We found that PLZF-RARα was only transiently expressed in nontumorigenic 32DCl3 cells after retroviral infection, suggesting a highly toxic or growth suppressive effect (Shaknovich and Licht, unpublished data). Pelicci’s group found that PLZF-RARα could be stably expressed in HL60, TF1, and U937 leukemic cells and blocks their differentiation in response to a number of chemical inducers.349 The transformed state of these cells might have allowed them to escape the potent growth-suppressive effects of PLZF-RARα. Consistent with the above results, PLZF-RARα, unlike PML-RARα, failed to increase the sensitivity of transduced cells to ATRA-mediated differentiation. HL60 cells lacking wild-type RARα transduced with PML-RARα showed increased expression of RARα target genes, whereas PLZF-RARα–transduced cells did not. Furthermore, whereas reintroduction of wild-type RARα or PML-RARα into the mutant HL-60 cells fully restored the ability of the cells to differentiate, as measured by the expression of leukocyte integrins, PLZF-RARα was only partially able to induce their expression. This information suggests that the differences in induction of endogenous genes by PLZF-RARα and PML-RARα are important for the ATRA-resistant clinical phenotype of t(11;17)(q23;q21)-associated APL. More physiologically relevant data indicate that PLZF-RARα on its own may not fully account for the ATRA resistance of t(11;17)(q23;q21) APL. Marrow progenitor cells infected with a PLZF-RARα retrovirus are able to be serially passaged ex vivo and displayed a primitive hematopoietic phenotype. Upon treatment of these cells with 10−7mol/L ATRA, proliferation ceased and differentiation ensued.336 

The role of the PLZF-RARα protein in leukemogenesis was further explored in a transgenic model using the cathepsin G promoter.299 These mice developed a chronic myeloid leukemia (CML)-like syndrome rather than APL. As in the case of the PML-RARα model, disease developed after a preleukemic phase, suggesting that secondary mutations are required for transformation. Acute leukemia developed more rapidly in these mice compared with the PML-RARα transgenics, suggesting that PLZF-RARα may be more oncogenic. Unlike PML-RARα transgenic mice, the PLZF-RARα mice did not achieve complete remission after ATRA treatment at a 10−6 mol/L dose, although they did show some evidence of myeloid differentiation. PLZF-RARα transgenic mouse leukemia cells treated with ATRA readily differentiated ex vivo, whereas in vivo, PLZF-RARα mice required a higher dose of ATRA than PML-RARα mice to induce short remissions.299 Thus, animals harboring PLZF-RARα were not absolutely insensitive to ATRA, suggesting that PLZF-RARα does not completely block ATRA-induced differentiation. The relative insensitivity of the disease in vivo correlates with the impaired ability of PLZF-RARα to transactivate in vitro, likely due to binding of corepressors even in the presence of ligand. This notion was further confirmed by the fact that the histone deacetylase inhibitor TSA in combination with ATRA synergistically inhibited growth and induced differentiation of PLZF-RARα harboring mouse leukemic cells. Therefore, the clinical phenotype of t(11;17)(q23;q21) APL might partly be due to the inability to achieve and sustain sufficiently high levels of ATRA required to stimulate the PLZF-RARα fusion product (Table5). A combination of ATRA and sodium butyrate, the latter already in clinical trial, might alleviate this situation.412,413 These animal experiments are still imperfect models of APL. PLZF-RARα mice, unlike PML-RARα mice, did not accumulate promyelocytes,299,342 although transgenic mice expressing PML-RARα under the cathepsin G promoter did not develop true APL either.341,342 In addition, the animal model does not explain the poor response of these patients to chemotherapy and the resistance of fresh t(11;17)(q23;q21) APL cells to high doses of ATRA in vitro.360,361,414 This information implicates another oncogenic lesion, potentially the reciprocal RARα-PLZF protein in the aggressive nature of this form of APL. This notion is supported by recent studies with fresh APL blasts of a patient with t(11;17)(q23;q21) APL.414 Neither ATRA nor arsenic treatment of these cells led to differentiation or apoptosis of the APL cells. Although arsenic can eliminate the microspeckled pattern of PML-RARα expression in APL associated with t(15;17), arsenic did not affect the punctate nuclear appearance of PLZF-RARα. ATRA treatment led to the degradation of PLZF-RARα, theoretically lifting the block to induction of RAR target genes, yet did not induce differentiation, suggesting that the reciprocal product offered a second oncogenic lesion.

In the case of the t(11;17)(q23;q21) APL variant, the reciprocal transcript encoding RARα-PLZF that yields a protein containing the last seven zinc fingers of PLZF fused to the A-domain of RARα361,362 (Fig 6) is consistently expressed. In contrast, the RARα-PML transcript of t(15;17) APL is absent in a significant number of cases.5 These seven zinc fingers can bind to the artificial PLZF binding site derived from PCR-based site selection as well as a site derived from selection from a human CpG island library.415 RARα-PLZF demonstrates properties that may be critical to the disruption of transcriptional and nuclear regulatory events. Whereas PLZF represses gene transcription through its cognate binding site, RARα-PLZF activates transcription through this site.394 Whereas PLZF is a growth suppressor and inhibits expression of the cyclin A2 gene, RARα-PLZF activates transcription, activates expression of cyclin A2 in an adhesion independent manner in 3T3 cells,406 and enhances cell growth (Yeyati et al, unpublished data). Hence, t(11;17)(q23;q21) may be an ATRA and chemotherapy-resistant disease due to the presence of two oncogenes working through different mechanisms, PLZF-RARα blocks retinoid-mediated activation of genes critical for myeloid differentiation. RARα-PLZF may activate cell cycle regulators such as cyclin A, accelerate cell growth, and block the antiproliferative effects of retinoid treatment. This notion is supported by the finding that mice harboring the RARα-PLZF protein develop a myeloproliferative syndrome. Whether these mice or the progeny of RARα-PLZF/PLZF-RARα crosses will develop leukemia is under study.416 

Finally, RARα-PLZF might also function through interference with RARα-mediated signaling. Inappropriate expression of this fusion protein, containing part of the AF1 activation domain of RARα, may act competitively with wild-type RARα for limiting cofactors. Similarly, a truncated form of RARγ containing the AF1 domain inhibited the ability of wild-type RARγ to activate transcription417 and transformion of keratinocytes, blocking their ability to differentiate. This supports the idea that a reciprocal RAR-X fusion protein could have an independent oncogenic effect.

In t(5;17)-associated APL, RARα is translocated to a region on chromosome 5q35 encoding the ubiquitously expressed and evolutionarily conserved nucleophosmin gene (NPM).418-420 NPM, also known as B23, numatrin, and NO38, was initially isolated as a nucleolar phosphoprotein in hepatoma cells.421-423 The human NPM gene spans 25 kb, consists of 12 exons,424 and has a promoter region consistent with those of housekeeping genes.425,426Alternative splicing yields two major isoforms: NPMB23.1(294 amino acids) and NPMB23.2 (257 amino acids), differing in their C-terminal region.420,427-429 Major structural features of NPM include two Asp/Glu-rich acidic domains, which may serve as binding sites for basic regions of other proteins; a bipartite nuclear localization signal (NLS); a metal binding motif; an ATP binding site; phosphorylation sites for cdc2 kinase and casein kinase II; and a binding site for proteins that contain nucleolar localization signals420,430-440 (Fig 9). The C-terminal region of the 294 amino acid NPMB23.1 isoform is also involved in nucleic acid binding and stimulation of DNA polymerase activity.441-444 Both isoforms reversibly multimerize to a hexameric state445 via the N-terminal domain.446 

NPM is localized most prominently to areas of the nucleolus associated with ribonucleoprotein (RNP) processing.235,237,447-449 NPM binds to nucleic acids, altering their conformation, an effect that could facilitate binding of ribosomal proteins to rRNA.237 NPM also copurifies with proteins required for DNA replication.443,450-452 In addition, NPM, particularly at the N-terminal region, is highly homologous to the evolutionarily conserved protein nucleoplasmin, a factor with chaperone activity that is involved in chromatin/nucleosome assembly.453,454 NPM also functions as part of a transport system used by ribosomal precursors to shuttle between the cytoplasm and nucleolus.236,440,455 NPM interacts with nonribosomal proteins via basic sequences (NoLS), assisting in transport to the nucleolus.436,455 456 

NPM levels are increased in proliferating cells457,458 and hypertrophic tissue to even higher levels in malignant cells,420,459,460 including leukemic blasts.461The increase in NPM expression may just be the consequence of increased requirements for ribosomal precursors. However, engineered overexpression of NPM in 3T3 cells yielded a transformed phenotype.461 One explanation for this could be that NPM binds to the tumor suppressor IRF-1 and inhibits its ability to activate genes that mediate the antiproliferative effect of IFN.461 Thus, NPM could behave as an antitumor suppressor.

NPM binds to transcription factor YY1, which is involved in cell growth and differentiation, changing it from a transcriptional repressor to an activator.462,463 YY1 binds to the NPM enhancer, possibly constituting a feedback mechanism.428 ATRA-induced differentiation of HL60 cells, but not growth arrest by serum withdrawal, resulted in downregulation of NPM.464 When cellular NPM levels were decreased by an antisense oligonucleotide, there was potentiation of the ATRA-induced differentiation.464 Growth-suppressive IRF-1 is upregulated by ATRA during myeloid differentiation, in opposition to the effect of NPM.115 These results support a possible role for NPM in the control of cellular growth and differentiation and hint at involvement in retinoid and IFN pathway regulatory cross-talk.

NPM undergoes dynamic changes over the course of the cell cycle. Expression peaks at S or G2 phase and is minimal in cells at G0.460,465-467 This pattern of expression might be related to the fact that NPMB23.1 specifically stimulates the activity of DNA polymerase α.444Alternatively, this may be a reflection of metabolic demands. In addition, NPM undergoes cdc2 threonine phosphorylation in G2/M.468,469 During M-phase progression, NPM associates with perichromosal regions and prenucleolar bodies, thus functionally linking the processes of nucleolar disassembly to mitotic chromosome condensation.469 The fact that NPM is intimately involved in events taking place at the G2/M regulatory point also underlines the strong relation between this protein and cellular proliferation.

NPM is preferentially regulated during apoptosis and cell damage. Apoptotic prostatic cells have hypophosphorylated NPM due to decreased CK II activity. A protease specific for unphosphorylated NPM then degrades the protein.470,471 NPM undergoes ADP-ribosylation after cells are exposed to x-rays, suggesting a role for NPM in DNA repair.472 NPM reversibly delocalizes from the nucleolus to the nucleoplasm235,473 when cells are exposed to conditions that discourage DNA or RNA synthesis or encourage terminal cell division, including stationary growth, serum starvation, hyperthermia, chemotherapeutic drugs, or ATRA (in HL-60 cells).235 473-483 This may reflect either decreased shuttling due to diminished metabolic requirements or a controlling role for NPM in regulating the cessation of cell growth. The dynamic response of NPM after various cell stresses resembles the reorganization of the PML-containing nuclear body under such conditions, indicating that both proteins may measure or control cell homeostatic processes.

NPM is fused to genes other then RARα in hematologic malignancies, as in the t(2;5)(p23;q35) translocation found in Ki-1+anaplastic large-cell lymphoma (ALCL).425,484-495 In this situation, NPM is linked to ALK, a gene encoding a membrane spanning tyrosine kinase425 normally not expressed in lymphoid tissue.496 The resulting protein contains the N-terminal oligomerization domain of NPM intracytoplasmic to the tyrosine kinase domain of ALK.424,497 As a result, the ubquitously expressed NPM gene drives the expression of an aberrant tryosine kinase, which can multimerize, yielding constitutive kinase activity, and aberrant oncogenic signaling.490,495,498-502 In the t(3;5)(q25.1;q34) translocation, found in myelodysplasia and M6-AML, a larger 175 amino acid portion of the NPM gene is linked to the MLF1 gene encoding an abundant cytoplasmic protein of unknown function.424 503 The resulting NPM-MLF fusion aberrantly places the MLF moiety in the nucleus and nucleolus. This fusion protein might also block normal NPM function, disrupting normal cellular growth control mechanisms. A similar mechanism could be at play in the case of NPM-RARα.

The t(5;17)(q35;q21) translocation was first described in a 2-year-old girl with APL504 who achieved cytogenetic remission after treatment with ATRA and chemotherapy. Blasts from this index patient, when thawed and treated with ATRA, differentiated into mature granulocytes.505 Subsequently, at least two other cases were described,418,506 one of whom has had a prolonged survival after ATRA and bone marrow transplantation.384Like the t(11;17)(q23;q21) APL syndrome, t(5;17) patients present with atypical morphological features, but unlike PLZF-associated APL, these patients are sensitive to ATRA therapy. This gene rearrangement joins the NPM gene 5′ to exon 3 of RARα in a manner similar to the other forms of APL.418 505 From a cDNA library derived from the marrow of the index patient, two fusion cDNAs were isolated. In the NPMS-RAR (520 aa), NPM contributes its oligomerization, metal binding, and nucleoplasmin homology domains as well as its highly active promoter. NPML-RARα (563aa) harbors an additional sequence of unclear origin, possibly from the noncoding region of NPM (Fig 9).

Like PML-RARα and PLZF-RARα, the NPM-RARα fusion acts as a ligand-dependent transcriptional activator when coexpressed with reporter genes containing RAREs,419 although it is not yet known whether NPM-RARα can act as a dominant negative inhibitor of wild-type RARα. This is likely, because the NPM moiety of the fusion, like the PML-and PLZF proteins, contains a multimerization domain and hence could sequester RXR and other cofactors from wild-type RARα. NPM-RARα has biological effects in cell culture similar to PML-RARα and PLZF-RARα.349 Its engineered expression in U937 cells blocked monocytic differentiation in response to vitamin D3and TGFβ.507 This furthers the notion that all of the RARα fusion proteins elicit the APL phenotype through disruption of nuclear receptor signaling. This was supported by the finding that PML-RARα, PLZF-RARα, and NPM-RARα could enhance the proliferation of primitive marrow progenitor cells after retrovirus-mediated gene transfer. Treatment of these cells with ATRA induced differentiation and inhibited cell growth.336 A transgenic model of t(5;17)-APL was recently created using the cathepsin G promoter. These mice developed an APL-like syndrome after a latent period, and blasts derived from these animals were ATRA sensitive.508 The response of this disease to ATRA may be mediated in part by downregulation of transcription of the NPM-RARα fusion, akin to the decline of NPM levels in differentiating HL60 cells.464This would relieve the putative dominant negative effect of the fusion protein.

NPM-RARα is expressed in a microspeckled pattern throughout the nucleus, similar to PML-RARα and PLZF-RARα.384 In this regard, NPM-RARα probably does not affect PML function, because PML was expressed in the wild-type nuclear body configuration in HeLa cells engineered to overexpress NPM-RARα, whereas NPM-RARα was expressed in a diffuse nuclear pattern234 and APL blasts from a t(5;17) patient exhibited a normal NB configuration.384Furthermore, NPM did not interact with PML in vitro. Interestingly, these experiments supply additional evidence that disruption of at least the PML component of the nuclear body is not required for the pathogenesis of APL.234 However, PLZF was delocalized in t(5;17) APL cells in a microspeckled pattern distinct from its wild-type distribution in normal marrow progenitor cells.384 This finding further supports a broad role for the PLZF protein in the pathogenesis of APL.

A reciprocal RARα-NPM mRNA was identified in the index t(5;17) patient, leading to fusion of the A domain of RARα to the acidic domains, NLS, and the rest of the C-terminus of NPM419 (Fig9). This molecule could associate with wild-type NPM through C-terminal sequences; however, it is unknown whether it interferes with any NPM function such as nucleolar-cytoplasmatic shuttling or apoptosis. RARα-NPM may bring the RARα activation domain in contact with NPM transcriptional targets such as YY1 and IRF-1, altering NPM-dependent transcriptional modulation. Lastly, similar to the other reciprocal fusion proteins, the ectopic expression of the RARα activation domain could interfere with wild-type RARα function by competition for AF1 cofactors.

The gene encoding the NuMA protein is the newest reported fusion partner of RARα in APL and the first mitotic apparatus protein found to be genetically rearranged in a human malignancy.509 NuMA is a large, highly abundant, conserved, and ubiquitously expressed protein that is intimately involved in the completion of mitosis and reformation of nuclei in the postmitotic daughter cells. NuMA is also a structural unit of the interphase nucleus.510-515 NuMA was first identified as an insoluble nonhistone chromatin-associated protein distributed diffusely throughout the interphase nucleus and displays a remarkable crescent-shaped mitotic staining pattern.516 517 

The NuMA gene, located on chromosome 11q13,518 encodes a highly conserved protein of 2,115 amino acids with a molecular weight of approximately 230 kD and by alternative splicing yields 1776aa and 1763aa proteins.519,520 The NuMA protein is divided into two globular domains at either end of the protein, with a central coiled region of 1485 aa515,521-523(Fig 10). The coiled motifs likely mediate protein homoassociation and heteroassociation.521,524 Little is known about the N-terminal globular region, although its absence impairs nuclear reformation after mitosis.522,525 The C-terminal region contains basic sequences, motifs for phosphorylation by cdc2 and other kinases,521,526 and sequences that confer localization to the nucleus (NLS) and mitotic spindle.520-522,525,527 528 

Fig. 10.

Functional domains of the NuMA protein and structure of the NuMA-RAR protein generated in t(11q13;17)-APL. As in the other fusion proteins of APL, an oligomerization domain contributed in this case from NuMA is linked to RAR. It is uncertain if a reciprocal protein is generated.

Fig. 10.

Functional domains of the NuMA protein and structure of the NuMA-RAR protein generated in t(11q13;17)-APL. As in the other fusion proteins of APL, an oligomerization domain contributed in this case from NuMA is linked to RAR. It is uncertain if a reciprocal protein is generated.

Close modal
Fig. 11.

Three axes to be investigated in understanding the pathogenesis of APL.

Fig. 11.

Three axes to be investigated in understanding the pathogenesis of APL.

Close modal

There is experimental evidence to support a role for NuMA in mitosis, apoptosis, and interphase nuclear matrix functions. It can participate in these diverse processes due to complex regulatory posttranslational modifications across the cell cycle. For example, NuMA is phosphorylated by the cdc2/cyclin B-regulatory kinase at the initiation of mitosis526,529 and associates with the spindle microtubules. A number of experiments demonstrated that NuMA is essential in forming convergent mitotic spindles and organizing and stabilizing parallel arrays of microtubules.510,511,525,527,530-535 NuMA may be required for the proper organization of newly forming daughter nuclei as cell division ends,511,517,527,531,532,536 forming a pathway for proteins to transit to daughter nuclei.515 Towards the end of mitosis, NuMA undergoes proteolytic cleavage and dephosphorylation.526 529 Furthermore, as cells progress towards G1, the remaining NuMA reverts to an insoluble form, yielding a fibrous network that may play a structural role during interphase. NuMA, like NPM, is an important target for regulation at the G2/M boundary as well as during postmitotic reorganization.

In interphase cells, immunofluorescence staining with anti-NuMA antibodies shows diffuse and/or speckled nuclear patterns.511,517,530,537 NuMA associates with a subset of nuclear core filaments around which proteins and nucleic acids coalesce to form the nuclear matrix.538 PML, NPM, and probably PLZF associate with the nuclear matrix.191,197,216,460,467 Other roles for NuMA were proposed based on coprecipitation with snRNPs and association with splicing complexes.539 In addition, NuMA specifically attaches to DNA matrix attachment regions (MAR), which are important for chromatin compaction and isolation of transcriptionally active loops of DNA.540 541 NuMA may thus provide structural support for RNA processing pathways, organize chromatin condensation, and participate in the regulation of transcriptional units.

NuMA may play a role in apoptosis and is an early target for proteolysis by caspase-3 and caspase-6,542 yielding an approximately 180-kD NuMA proteolytic product,526,543,544lacking the C-terminal globular effector domain after several different apoptotic stimuli.544-547 This proteolytic product could act as a dominant negative, disrupting normal nuclear structure. Alternatively, release of NuMA from the DNA matrix attachment regions may facilitate the DNA fragmentation characteristic of apoptosis.546 However, in some of these experimental models, there was no correlation between nuclear fragmentation and NuMA proteolysis,544 suggesting that NuMA may be specifically targeted to prevent dying cells from proceeding through M-phase. In summary, NuMA would appear to be a structural component of the cell that responds to cell cycle signals on cue rather than a controlling factor in cell proliferation. It is unclear whether inhibition of NuMA function might contribute to oncogenesis. In fact, many NuMA mutants seriously disrupt mitosis and would not be expected to be compatible with normal cell growth and proliferation.

The NuMA-RARα fusion protein was first described in a 6-month-old boy with morphologically diagnosed APL harboring a translocation t(11;17)(q13;q21).509 The patient had a complete remission after ATRA therapy and was disease free at 24+ months after a bone marrow transplant.509 The t(11;17)(q13;q21) results in a 2286 aa protein predicted to consist of 1883 amino acids of NuMA, including the N-terminal globular and coiled-coil domains of NuMA fused to RARα domains B through F, as in all the other RARα fusion proteins548 (Fig 10). NuMA-RARα localized to sheetlike nuclear aggregates in the patient’s leukemic cells, but NB structure and PML staining were unperturbed. Introduction of C-terminal deletion mutants of NuMA into cells completely disrupts mitosis,522 525 and it is surprising that the existence of NuMA-RARα is even compatible with cell division. Perhaps the RARα moiety mislocalizes the mutant NuMA from the daughter nuclei reforming after mitosis, allowing the remaining wild-type NuMA to perform its function. In the nucleus, the fusion may, in a dominant negative manner, interfere with regulatory events during interphase or G2. NuMA-RARα might also compete with wild-type NuMA for caspases, interfering with apoptosis.

The most likely mechanism of action of the NuMA-RARα is interference with nuclear receptor function. Although no functional data for the NuMA-RARα fusion are yet available, it is probable that, like the other RARα fusion proteins, NuMA-RARα is a dominant negative retinoid receptor with diminished intrinsic transcriptional activity. A fundamental common denominator among APL fusion proteins is their ability to form higher order complexes. Like PML, PLZF, and NPM, NuMA can multimerize.521,524 NuMA-RARα might thus sequester RARα partner proteins or have aberrant affinity for nuclear receptor coactivators or corepressors. Like PML, NuMA is a component of the nuclear matrix and NuMA-RARα might further inhibit RARα function by confinement of RARα cofactors in a subnuclear compartment (such as the MAR) apart from wild-type RARα. Another common theme among RARα fusion proteins is their downregulation when treated with ATRA. Similarly, NuMA-RARα might decrease as t(11;17)(q13;q21) APL cells are induced to terminally differentiate by ATRA. This is consistent with the fact that normal neutrophils do not express NuMA.547 Finally, it is unknown whether a reciprocal RARα-NuMA transcript is expressed in this disease. In fact, there might be selection against this protein as it could represent a detriment to mitosis.

There are three axes to be investigated in understanding the pathogenesis of APL (Fig 11).

First, it is clear that certain RARα target genes must be activated (or repressed) for myeloid development to proceed. In all APL patients, one allele of RARα is disrupted and fused to the partner gene, N. All of the N genes encode a protein able to multimerize. The multimerization of the N-RARα fusion protein may be a key factor in interfering with normal ATRA-mediated signaling and sequestering cofactors. Recent evidence shows that PML-RARα and PLZF-RARα have abnormally high affinities for corepressors. Because PML alone does not seem to bind these factors, it appears that fusion of RARα to the PML moiety effects an allosteric change in the entire molecule, causing the fusion protein to retain the corepressors at physiological concentrations of ATRA. The structural basis of this change is unknown. NPM and NuMA might have the same effect on RARα, and this should be investigated. In contrast, PLZF binds the corepressors even in the presence of ATRA. The net result is that, at physiological levels of ATRA, the fusion receptors do not activate the critical genes for myeloid differentiation and only can do so with pharmacological doses. In the case of PLZF-RARα, even the usual pharmacologic dose of ATRA is insufficient for full activation of the RARα axis. The use of of 10−5 mol/L ATRA and/or addition of HDAC inhibitors are required to allow retinoic acid signaling to proceed.

Second, the N gene is disrupted with only one normal allele remaining. Loss of N gene dose and/or function might play a role in the pathogenesis of APL. The N-RARα fusion could sequester the normal N product, altering its role in growth control. PML-RARα delocalizes both PML and PLZF from nuclear bodies, potentially altering their function. Whether NPM-RARα and NuMA-RARα affect the function of the wild-type proteins is unknown.

Third, the reciprocal RARα-N fusion gene could play a role in the pathogenesis or clinical phenotype of the APL syndrome. The N-RARα fusion, in addition to altering RARα function, could act to as a second dominant negative protein to inhibit the function of the N protein. In the cases of t(11;17)(q23;q21)-associated APL, the RARα-PLZF gene product could both interfere with normal PLZF function and have novel gains of function. Hence, many questions remain, including the following:

(1) What are the critical RARα target genes required for myeloid differentiation that are inhibited in their expression by the N-RARα protein?

(2) What are the molecular details of the corepressor complex bound to N-RARα that inhibit the activity of the fusion protein? Are all the corepressors directly bound or are a chain of protein-protein interactions required? Are coactivators also sequestered by the N-RARα fusions? Will manipulation of coactivators (histone acetylases) as well as corepressor histone deacetylase factors play a role in restoration of normal myeloid differentiation and the treatment of APL?

(3) Is the NB an active organelle or an intranuclear storage site? What is significance of the PML and PLZF association with the NB? Is there a role for NB disruption in the pathogenesis of APL?

(4) What is the mechanism of action of the PML protein in transcription, apoptosis, and growth control?

(5) What is the role of PLZF in hematopoiesis and what are its target genes?

(6) How do RARα fusion proteins transform cells? Do the fusion proteins alter expression of regulators of apoptosis? Are the RARα fusion proteins dominant negative inhibitors of PML or PLZF? Will APL develop both in PML or PLZF null animals?

(7) How does arsenic cause the apoptosis of APL cells? Is arsenic response dependent on the presence of N-RARα fusion proteins or is it reflective of the particular state of differentiation of the APL cell?

(8) Do RARα-N reciprocal fusion proteins cooperate with N-RARα proteins to induce leukemia?

(9) Can other molecular pathways, such as caspases or IFN mediators, be exploited as avenues for future therapies of APL?

(10) Are there molecular interactions between PML and NPM or NuMA?

The study of the molecular pathogenesis of APL is at the forefront of the application of molecular biology to clinical medicine, because this disease is the paradigm for successful differentiation therapy. The spectacular response of these patients to ATRA has underlined the importance of continued efforts to understand the basic biology of leukemia. We now understand that the RARα fusion genes of APL are the key to the cause and cure of this disease. Work to date has elucidated how the RARα fusion proteins may block differentiation and how ATRA can reverse this block and promote differentiation and death of the malignant clone. Translational studies using clinical samples have highlighted how retinoic acid resistance can occur in patients who develop secondary mutations in the PML-RARα fusion gene. New work has led to a greater understanding of how arsenic, a reactivated agent of the cancer armamentarium, may promote death of neoplastic cells. The occurrence of naturally resistant forms of APL such as that associated with rearrangement of the PLZF gene indicates that molecular heterogeneity can occur in APL and that a definitive diagnosis of t(15;17)-APL must be made before ATRA can be used with confidence. The study of the resistant and sensitive forms of APL, characterized by rearrangement of the PML and PLZF genes, respectively, yielded an appreciation of the importance of transcriptional repression by histone deacetylation in the development of the disease. This led to the recent use of the deacetylase inhibitor sodium butyrate as a form of targeted transcription therapy in a patient with resistant APL.549With the development of animal and cell models of APL of sensitive and resistant forms of APL and the advent of more powerful technologies for gene discovery and cell biology, the next 5 years should offer continued insights leading to the development of more effective therapies for this fascinating disease as well as other forms of leukemia.

The authors thank S. Waxman, J. Gabrilove, G. Atweh, M. McConnell, and A. Zelent for review of the manuscript and C. Brechok for editorial assistance.

Supported by Grant No. R01 CA 59936 (J.D.L.) and American Cancer Society Award DHP 160 (J.D.L.). J.D.L. is a scholar of the Leukemia Society of America. A.M. is supported by Grant No. K08 CA73762.

1
Huang
M
Ye
Y
SR
C
Zhao
J
Gu
L
Cai
J
Zhao
L
Xie
J
Shen
Z
Wang
Z
All-Trans retinoic acid with or without low dose Ara-C in acute promyelocytic leukemia.
Chinese Med J
100
1987
949
2
Huang
M
Ye
Y
Chen
S
Chai
J
Lu
J
Lin
Z
Gu
L
Wang
Z
Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia.
Blood
72
1988
567
3
Warrell
RP
Jr
de The
H
Wang
ZY
Degos
L
Acute promyelocytic leukemia.
N Engl J Med
329
1993
177
4
Degos
L
Differentiation therapy of leukemia.
Leuk Lymphoma
13
1994
39
(suppl 1)
5
Grignani
F
Fagioli
M
Alcalay
M
Longo
L
Pandolfi
PP
Donti
E
Biondi
A
Lo Coco
F
Grignani
F
Pelicci
PG
Acute promyelocytic leukemia: from genetics to treatment.
Blood
83
1994
10
6
Tallman
MS
Differentiating therapy with all-trans retinoic acid in acute myeloid leukemia.
Leukemia
10
1996
12
(suppl 1)
7
Fenaux
P
Chomienne
C
Degos
L
Acute promyelocytic leukemia: Biology and treatment.
Semin Oncol
24
1997
92
8
Fenaux
P
Degos
L
Differentiation therapy for acute promyelocytic leukemia.
N Engl J Med
337
1997
1076
9
Rowley
J
Golumb
H
Dougherty
C
15/17 translocation a consistant chromsomal change in acute promyelocytic leukemia.
Lancet
1
1977
549
10
de The
H
Chomienne
C
Lanotte
M
Degos
L
Dejean
A
The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor alpha gene to a novel transcribed locus.
Nature
347
1990
558
11
Chang
KS
Stass
SA
Chu
DT
Deaven
LL
Trujillo
JM
Freireich
EJ
Characterization of a fusion cDNA (RARA/myl) transcribed from the t(15;17) translocation breakpoint in acute promyelocytic leukemia.
Mol Cell Biol
12
1992
800
12
Goddard
A
Borrow
J
Freemont
P
Solomon
E
Characterization of a zinc finger gene disrupted by the t(15;17) in acute promyelocytic leukemia.
Science
254
1991
1371
13
Kakizuka
A
Miller
WJ
Umesono
K
Warrell
RJ
Frankel
SR
Murty
VV
Dmitrovsky
E
Evans
RM
Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RAR alpha with a novel putative transcription factor, PML.
Cell
66
1991
663
14
Alcalay
M
Zangrilli
D
Pandolfi
PP
Longo
L
Mencarelli
A
Giacomucci
A
Rocchi
M
Biondi
A
Rambaldi
A
Lo Coco
F
Diverio
D
Grignani
F
Pelicci
PG
Translocation breakpoint of acute promyelocytic leukemia lies within the retinoic acid receptor alpha locus.
Proc Natl Acad Sci USA
88
1991
1977
15
Pandolfi
PP
Grignani
F
Alcalay
M
Mencarelli
A
Biondi
A
LoCoco
F
Grignani
F
Pelicci
PG
Structure and origin of the acute promyelocytic leukemia myl/RAR alpha cDNA and characterization of its retinoid-binding and transactivation properties.
Oncogene
6
1991
1285
16
Soignet
SL
Maslak
P
Wang
ZG
Jhanwar
S
Calleja
E
Dardashti
LJ
Corso
D
DeBlasio
A
Gabrilove
J
Scheinberg
DA
Pandolfi
PP
Warrell
RP
Jr
Complete remission after treatment of acute promyelocytic leukemia with arsenic trioxide.
N Engl J Med
339
1998
1341
17
Gallagher
RE
Arsenic—New life for an old potion.
N Engl J Med
339
1998
1389
18
Wolbach
S
Howe
P
Tissue changes following deprivation of fat-soluble A vitamin.
J Exp Med
42
1925
753
19
Hodges
R
Sauberlich
H
Canham
J
Wallace
D
Rucker
R
Meja
L
Monhanram
M
Hematopoietic studies in Vitamin A deficiency.
Am J Clin Nutr
31
1977
876
20
Douer
D
Koeffler
HP
Retinoic acid. Inhibition of the clonal growth of human myeloid leukemia cells.
J Clin Invest
69
1982
277
21
Gratas
C
Menot
ML
Dresch
C
Chomienne
C
Retinoid acid supports granulocytic but not erythroid differentiation of myeloid progenitors in normal bone marrow cells.
Leukemia
7
1993
1156
22
Breitman
T
Collins
S
Selonick
S
Induction of differentiation of the human promyelocytic leukemia cell line (HL60) by retinoic acid.
Proc Natl Acad Sci USA
77
1980
2936
23
Breitman
T
Collins
SJ
Keene
B
Terminal differentiation of human promeylocytic cells in culture in response to retinoic acid.
Blood
57
1981
1000
24
Evans
RM
The steroid and thyroid hormone receptor superfamily.
Science
240
1988
889
25
Mangelsdorf
DJ
Evans
RM
The RXR heterodimers and orphan receptors.
Cell
83
1995
841
26
Glass
CK
DiRenzo
J
Kurokawa
R
Han
ZH
Regulation of gene expression by retinoic acid receptors.
DNA Cell Biol
10
1991
623
27
de The
H
Marchio
A
Tiollais
P
Dejean
A
Differential expression and ligand regulation of the retinoic acid receptor alpha and beta genes.
EMBO J
8
1989
429
28
Gallagher
RE
Said
F
Pua
I
Papenhausen
PR
Paietta
E
Wiernik
PH
Expression of retinoic acid receptor-alpha mRNA in human leukemia cells with variable responsiveness to retinoic acid.
Leukemia
3
1989
789
29
Largman
C
Detmer
K
Corral
JC
Hack
FM
Lawrence
HJ
Expression of retinoic acid receptor alpha mRNA in human leukemia cells.
Blood
74
1989
99
30
Chambon
P
A decade of molecular biology of retinoic acid receptors.
FASEB J
10
1996
940
31
Leroy
P
Krust
A
Zelent
A
Mendelsohn
C
Garnier
J-M
Kastner
P
Dierich
A
Chambon
P
Multiple isoforms of the mouse retinoic acid receptor α are generated by alternative splicing and differential induction by retinoic acid.
EMBO J
10
1991
59
32
Leroy
P
Naksharti
H
Chambon
P
Mouse retinoic acid receptor a2 isoform is transcribed from a promoter that contains a retinoic acid response element.
Proc Natl Acad Sci USA
88
1991
10138
33
de Thé
H
Vivanco-Ruiz
MM
Tiollais
P
Stunnenberg
H
Dejean
A
Identification of a retinoic acid responsive element in the retinoic acid receptor beta gene.
Nature
343
1990
177
34
Sucov
HM
Murakami
KK
Evans
RM
Characterization of an autoregulated response element in the mouse retinoic acid receptor type β gene.
Proc Natl Acad Sci USA
87
1990
5392
35
Lehmann
JM
Zhang
XK
Pfahl
M
RAR gamma 2 expression is regulated through a retinoic acid response element embedded in Sp1 sites.
Mol Cell Biol
12
1992
2976
36
Umesono
K
Murakami
KK
Thompson
CC
Evans
RM
Direct repeats as selective response elements for the thyroid hormone, retinoic acid, and vitamin D3 receptors.
Cell
65
1991
1255
37
Naar
AM
Boutin
JM
Lipkin
SM
Yu
VC
Holloway
JM
Glass
CK
Rosenfeld
MG
The orientation and spacing of core DNA-binding motifs dictate selective transcriptional responses to three nuclear receptors.
Cell
65
1991
1267
38
Gudas
LJ
Retinoids and vertebrate development.
J Biol Chem
269
1994
15399
39
Zechel
C
Shen
XQ
Chambon
P
Gronemeyer
H
Dimerization interfaces formed between the DNA binding domains determine the cooperative binding of RXR/RAR and RXR/TR heterodimers to DR5 and DR4 elements.
EMBO J
13
1994
1414
40
Perlmann
T
Umesono
K
Rangarajan
PN
Forman
BM
Evans
RM
Two distinct dimerization interfaces differentially modulate target gene specificity of nuclear hormone receptors.
Mol Endocrinol
10
1996
958
41
Mangelsdorf
DJ
Borgmeyer
U
Heyman
RA
Zhou
JY
Ong
ES
Oro
AE
Kakizuka
A
Evans
RM
Characterization of three RXR genes that mediate the action of 9-cis retinoic acid.
Genes Dev
6
1992
329
42
Nagpal
S
Saunders
M
Kastner
P
Durand
B
Naksharti
H
Chambon
P
Promoter context- and response element-dependent specificity of the transcriptional activation and modulating functions of retinoic acid receptors.
Cell
70
1992
1007
43
Nagpal
S
Friant
S
Nakshatri
H
Chambon
P
RARs and RXRs: Evidence for two autonomous transactivation functions (AF-1 and AF-2) and heterodimerization in vivo.
EMBO J
12
1993
2349
44
Chen
JD
Evans
RM
A transcriptional co-repressor that interacts with nuclear hormone receptors.
Nature
377
1995
454
45
Horlein
AJ
Naar
AM
Heinzel
T
Torchia
J
Gloss
B
Kurokawa
R
Ryan
A
Kamei
Y
Soderstrom
M
Glass
CK
Rosenfeld
MG
Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor.
Nature
377
1995
397
46
Pazin
M
Kadonaga
J
What’s up and down with histone deacetylation and transcription.
Cell
89
1997
325
47
Bourguet
W
Ruff
M
Bonnier
D
Granger
F
Boeglin
M
Chambon
P
Moras
D
Gronemeyer
H
Purification, functional characterization, and crystallization of the ligand binding domain of the retinoid X receptor.
Protein Expr Purif
6
1995
604
48
Bourguet
W
Ruff
M
Chambon
P
Gronemeyer
H
Moras
D
Crystal structure of the ligand-binding domain of the human nuclear receptor RXR-alpha.
Nature
375
1995
377
49
Renaud
JP
Rochel
N
Ruff
M
Vivat
V
Chambon
P
Gronemeyer
H
Moras
D
Crystal structure of the RAR-gamma ligand-binding domain bound to all-trans retinoic acid.
Nature
378
1995
681
50
vom Baur
E
Zechel
C
Heery
D
Heine
MJ
Garnier
JM
Vivat
V
Le Douarin
B
Gronemeyer
H
Chambon
P
Losson
R
Differential ligand-dependent interactions between the AF-2 activating domain of nuclear receptors and the putative transcriptional intermediary factors mSUG1 and TIF1.
EMBO J
15
1996
110
51
LeDouarin
B
Zechel
C
Garnier
JM
Lutz
Y
Tora
L
Pierrat
P
Heery
D
Gronemeyer
H
Chambon
P
Losson
R
The N-terminal part of TIF1, a putative mediator of the ligand-dependent activation function (AF-2) of nuclear receptors, is fused to B-raf in the oncogenic protein T18.
EMBO J
14
1995
2020
52
Lee
JW
Ryan
F
Swaffield
JC
Johnston
SA
Moore
DD
Interaction of thyroid-hormone receptor with a conserved transcriptional mediator.
Nature
374
1995
91
53
Voegel
JJ
Heine
MJ
Zechel
C
Chambon
P
Gronemeyer
H
TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors.
EMBO J
15
1996
3667
54
Chen
H
Lin
RJ
Schiltz
RL
Chakravarti
D
Nash
A
Nagy
L
Privalsky
ML
Nakatani
Y
Evans
RM
Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300.
Cell
90
1997
569
55
Onate
SA
Tsai
SY
Tsai
MJ
O’Malley
BW
Sequence and characterization of a coactivator for the steroid hormone receptor superfamily.
Science
270
1995
1354
56
Mengus
G
May
M
Carre
L
Chambon
P
Davidson
I
Human TAF(II)135 potentiates transcriptional activation by the AF-2s of the retinoic acid, vitamin D3, and thyroid hormone receptors in mammalian cells.
Genes Dev
11
1997
1381
57
Yao
TP
Ku
G
Zhou
N
Scully
R
Livingston
DM
The nuclear hormone receptor coactivator SRC-1 is a specific target of p300.
Proc Natl Acad Sci USA
93
1996
10626
58
Kamei
Y
Xu
L
Heinzel
T
Torchia
J
Kurokawa
R
Gloss
B
Lin
S-C
Heyman
R
Rose
D
Glass
C
Rosenfeld
M
A CBP integrator complex mediates transcriptional activation and AP-1 inhibition by nuclear receptors.
Cell
85
1996
403
59
Chakravarti
D
LaMorte
VJ
Nelson
MC
Nakajima
T
Schulman
IG
Juguilon
H
Montminy
M
Evans
RM
Role of CBP/P300 in nuclear receptor signalling.
Nature
383
1996
99
60
Tjian
R
Maniatis
T
Transcriptional activation: A complex puzzle with few easy pieces.
Cell
77
1994
5
61
Fraser
RA
Heard
DJ
Adam
S
Lavigne
AC
Le Douarin
B
Tora
L
Losson
R
Rochette-Egly
C
Chambon
P
The putative cofactor TIF1alpha is a protein kinase that is hyperphosphorylated upon interaction with liganded nuclear receptors.
J Biol Chem
273
1998
16199
62
Fraser
RA
Rossignol
M
Heard
DJ
Egly
JM
Chambon
P
SUG1, a putative transcriptional mediator and subunit of the PA700 proteasome regulatory complex, is a DNA helicase.
J Biol Chem
272
1997
7122
63
Yang
XJ
Ogryzko
VV
Nishikawa
J
Howard
BH
Nakatani
Y
A p300/CBP-associated factor that competes with the adenoviral oncoprotein E1A.
Nature
382
1996
319
64
Ogryzko
VV
Schiltz
RL
Russanova
V
Howard
BH
Nakatani
Y
The transcriptional coactivators p300 and CBP are histone acetyltransferases.
Cell
87
1996
953
65
Bannister
AJ
Kouzarides
T
The CBP co-activator is a histone acetyltransferase.
Nature
384
1996
641
66
Blanco
JC
Minucci
S
Lu
J
Yang
XJ
Walker
KK
Chen
H
Evans
RM
Nakatani
Y
Ozato
K
The histone acetylase PCAF is a nuclear receptor coactivator.
Genes Dev
12
1998
1638
67
Wade
PA
Wolffe
AP
Histone acetyltransferases in control.
Curr Biol
7
1997
R82
68
Chen
JY
Clifford
J
Zusi
C
Starrett
J
Tortolani
D
Ostrowski
J
Reczek
PR
Chambon
P
Gronemeyer
H
Two distinct actions of retinoid-receptor ligands.
Nature
382
1996
819
69
Dawson
MI
Elstner
E
Kizaki
M
Chen
DL
Pakkala
S
Kerner
B
Koeffler
HP
Myeloid differentiation mediated through retinoic acid receptor/retinoic X receptor (RXR) not RXR/RXR pathway.
Blood
84
1994
446
70
Kizaki
M
Nakajima
H
Mori
S
Koike
T
Morikawa
M
Ohta
M
Saito
M
Koeffler
HP
Ikeda
Y
Novel retinoic acid, 9-cis retinoic acid, in combination with all-trans retinoic acid is an effective inducer of differentiation of retinoic acid-resistant HL-60 cells.
Blood
83
1994
3289
71
Benedetti
L
Grignani
F
Scicchitano
BM
Jetten
AM
Diverio
D
Lo
CF
Avvisati
G
Gambacorti
PC
Adamo
S
Levin
AA
Pelicci
PG
Nervi
C
Retinoid-induced differentiation of acute promyelocytic leukemia involves PML-RARalpha-mediated increase of type II transglutaminase.
Blood
87
1996
1939
72
Pfahl
M
Nuclear receptor/AP-1 interaction.
Endocr Rev
14
1993
651
73
Kamei
Y
Xu
L
Heinzel
T
Torchia
J
Kurokawa
R
Gloss
B
Lin
SC
Heyman
RA
Rose
DW
Glass
CK
Rosenfeld
MG
A CBP integrator complex mediates transcriptional activation and AP-1 inhibition by nuclear receptors.
Cell
85
1996
403
74
Horvai
AE
Xu
L
Korzus
E
Brard
G
Kalafus
D
Mullen
TM
Rose
DW
Rosenfeld
MG
Glass
CK
Nuclear integration of JAK/STAT and Ras/AP-1 signaling by CBP and p300.
Proc Natl Acad Sci USA
94
1997
1074
75
Janknecht
R
Hunter
T
Transcription. A growing coactivator network.
Nature
383
1996
22
76
Kizaki
M
Dawson
MI
Heyman
R
Elster
E
Morosetti
R
Pakkala
S
Chen
DL
Ueno
H
Chao
W
Morikawa
M
Ikeda
Y
Heber
D
Pfahl
M
Koeffler
HP
Effects of novel retinoid X receptor-selective ligands on myeloid leukemia differentiation and proliferation in vitro.
Blood
87
1996
1977
77
Collins
SJ
Robertson
KA
Mueller
L
Retinoic acid-induced granulocytic differentiation of HL-60 myeloid leukemia cells is mediated directly through the retinoic acid receptor (RAR-alpha).
Mol Cell Biol
10
1990
2154
78
Robertson
KA
Emami
B
Collins
SJ
Retinoic acid-resistant HL-60R cells harbor a point mutation in the retinoic acid receptor ligand-binding domain that confers dominant negative activity.
Blood
80
1992
1885
79
Damm
K
Heyman
RA
Umesono
K
Evans
RM
Functional inhibition of retinoic acid response by dominant negative retinoic acid receptor mutants.
Proc Natl Acad Sci USA
90
1993
2989
80
Robertson
KA
Emami
B
Mueller
L
Collins
SJ
Multiple members of the retinoic acid receptor family are capable of mediating the granulocytic differentiation of HL-60 cells.
Mol Cell Biol
12
1992
3743
81
Zelent
A
Zhu
J
Lanotte
M
Gallagher
R
Waxman
S
Heyworth
CM
Enver
T
Differential expression of retinoid receptors during multilineage differentiation of haemopoietic progentior cells-role of the RARα2 isoform in normal granulopoiesis and leukemia.
Blood
90
1997
44a
(abstr, supp 1)
82
Tsai
S
Bartelmez
S
Heyman
R
Damm
K
Evans
R
Collins
SJ
A mutated retinoic acid receptor-alpha exhibiting dominant-negative activity alters the lineage development of amultipotent hematopoietic cell line.
Genes Dev
6
1992
2258
83
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
84
Morosetti
R
Grignani
F
Liberatore
C
Pelicci
PG
Schiller
GJ
Kizaki
M
Bartram
CR
Miller
CW
Koeffler
HP
Infrequent alterations of the RAR alpha gene in acute myelogenous leukemias, retinoic acid-resistant acute promyelocytic leukemias, myelodysplastic syndromes, and cell lines.
Blood
87
1996
4399
85
Onodera
M
Kunisada
T
Nishikawa
S
Sakiyama
Y
Matsumoto
S
Nishikawa
S
Overexpression of retinoic acid receptor alpha suppresses myeloid cell differentiation at the promyelocyte stage.
Oncogene
11
1995
1291
86
Ptashne
M
How eukaryotic transcriptional activators work.
Nature
335
1988
683
87
Dey
A
Minucci
S
Ozato
K
Ligand-dependent occupancy of the retinoic acid receptor beta 2 promoter in vivo.
Mol Cell Biol
14
1994
8191
88
Frankel
SR
Eardley
A
Lauwers
G
Weiss
M
Warrell
RP
Jr
The “retinoic acid syndrome” in acute promyelocytic leukemia.
Ann Intern Med
117
1992
292
89
Barbui
T
Finazzi
G
Falanga
A
The impact of all-trans-retinoic acid on the coagulopathy of acute promyelocytic leukemia.
Blood
91
1998
3093
90
Vyas
RC
Frankel
SR
Agbor
P
Miller
WH
Jr
Warrell
RP
Jr
Hittelman
WN
Probing the pathobiology of response to all-trans retinoic acid in acute promyelocytic leukemia: Premature chromosome condensation/fluorescence in situ hybridization analysis.
Blood
87
1996
218
91
Glasser
L
Fiederlein
RL
Shamdas
GJ
Brothman
AR
Functional characteristics of in vivo induced neutrophils after differentiation therapy of acute promyelocytic leukemia with all-trans-retinoic acid.
Cancer
73
1994
1206
92
Miyauchi
J
Ohyashiki
K
Inatomi
Y
Toyama
K
Neutrophil secondary-granule deficiency as a hallmark of all-trans retinoic acid-induced differentiation of acute promyelocytic leukemia cells.
Blood
90
1997
803
93
Khanna-Gupta
A
Kolibaba
K
Zibello
TA
Berliner
N
NB4 cells show bilineage potential and an aberrant pattern of neutrophil secondary granule protein gene expression.
Blood
84
1994
294
94
Chomienne
C
Balitrand
N
Ballerini
P
Castaigne
S
de The
H
Degos
L
All-trans retinoic acid modulates the retinoic acid receptor-alpha in promyelocytic cells.
J Clin Invest
88
1991
2150
95
Yoshida
H
Kitamura
K
Tanaka
K
Omura
S
Miyazaki
T
Hacahiya
T
Ohno
R
Naoe
T
Accelerated degradation of PML-retinoic acid receptor-alpha (PML-RARA) oncoprotein by all-trans retinoic acid in acute promyelocytic leukemia: Possible role of the protesome pathway.
Cancer Res
56
1996
2945
96
Raelson
J
Nervi
C
Rosenauer
A
Benedetti
L
Monczak
Y
Pearson
M
Pelicci
P
Miller
J, WH
The PML/RARα oncoprotein is a direct molecular target of retinoic acid in acute promyelocytic leukemia cells.
Blood
88
1996
2826
97
Krumlauf
R
Hox genes in vertebrate development.
Cell
78
1994
191
98
Marshall
H
Morrison
A
Studer
M
Popperl
H
Krumlauf
R
Retinoids and Hox genes.
FASEB J
10
1996
969
99
Magli
MC
Barba
P
Celetti
A
De Vita
G
Cillo
C
Boncinelli
E
Coordinate regulation of HOX genes in human hematopoietic cells.
Proc Natl Acad Sci USA
88
1991
6348
100
Shen
WF
Detmer
K
Mathews
CH
Hack
FM
Morgan
DA
Largman
C
Lawrence
HJ
Modulation of homeobox gene expression alters the phenotype of human hematopoietic cell lines.
EMBO J
11
1992
983
101
Blatt
C
Lotem
J
Sachs
L
Inhibition of specific pathways of myeloid cell differentiation by an activated Hox-2.4 homeobox gene.
Cell Growth Differ
3
1992
671
102
Allen
JD
Adams
JM
Enforced expression of Hlx homeobox gene prompts myeloid cell maturation and altered adherence properties of T cells.
Blood
81
1993
3242
103
Nakamura
T
Largaespada
DA
Shaughnessy
JD
Jr
Jenkins
NA
Copeland
NG
Cooperative activation of Hoxa and Pbx1-related genes in murine myeloid leukaemias.
Nat Genet
12
1996
149
104
Giampaolo
A
Pelosi
E
Valtieri
M
Montesoro
E
Sterpetti
P
Samoggia
P
Camagna
A
Mastroberardino
G
Gabbianelli
M
Testa
U
Peschle
C
HOXB gene expression and function in differentiating purified hematopoietic progenitors.
Stem Cells
13
1995
90
105
Kamps
MP
Wright
DD
Oncoprotein E2A-Pbx1 immortalizes a myeloid progenitor in primary marrow cultures without abrogating its factor-dependence.
Oncogene
9
1994
3159
106
Borrow
J
Shearman
AM
Stanton
VP
Jr
Becher
R
Collins
T
Williams
AJ
Dube
I
Katz
F
Kwong
YL
Morris
C
Ohyashiki
K
Toyama
K
Rowley
J
Housman
DE
The t(7;11)(p15;p15) translocation in acute myeloid leukaemia fuses the genes for nucleoporin NUP98 and class I homeoprotein HOXA9.
Nat Genet
12
1996
159
107
Sauvageau
G
Thorsteindottir
U
Eaves
C
Lawrence
H
Largman
C
Lansdorp
P
Humphries
RK
Overexpression of HOXB4 in hematopoietic cells causes the selective expansion of more primative populations in vitro and in vivo.
Genes Dev
9
1995
1753
108
Marshall
H
Studer
M
Popperl
H
Aparicio
S
Kuroiwa
A
Brenner
S
Krumlauf
R
A conserved retinoic acid response element required for early expression of the homeobox gene Hoxb-1.
Nature
370
1994
567
109
Ogura
T
Evans
RM
A retinoic acid-triggered cascade of HOXB1 gene activation.
Proc Natl Acad Sci USA
92
1995
387
110
Langston
AW
Thompson
JR
Gudas
LJ
Retinoic acid-responsive enhancers located 3′ of the Hox A and Hox B homeobox gene clusters. Functional analysis.
J Biol Chem
272
1997
2167
111
Langston
AW
Gudas
LJ
Identification of a retinoic acid responsive enhancer 3′ of the murine homeobox gene Hox-1.6.
Mech Dev
38
1992
217
112
Frasch
M
Chen
X
Lufkin
T
Evolutionary-conserved enhancers direct region-specific expression of the murine Hoxa-1 and Hoxa-2 loci in both mice and Drosophila.
Development
121
1995
957
113
Morrison
A
Moroni
MC
Ariza-McNaughton
L
Krumlauf
R
Mavilio
F
In vitro and transgenic analysis of a human HOXD4 retinoid-responsive enhancer.
Development
122
1996
1895
114
Boylan
JF
Lufkin
T
Achkar
CC
Taneja
R
Chambon
P
Gudas
LJ
Targeted disruption of retinoic acid receptor alpha (RAR alpha) and RAR gamma results in receptor-specific alterations in retinoic acid-mediated differentiation and retinoic acid metabolism.
Mol Cell Biol
15
1995
843
115
Matikainen
S
Ronni
T
Hurme
M
Pine
R
Julkunen
I
Retinoic acid activates interferon regulatory factor-1 gene expression in myeloid cells.
Blood
88
1996
114
116
Harada
H
Kitakawa
M
Tanaka
N
Yamamoto
H
Harada
K
Ishihara
M
Taniguchi
T
Anti-oncogenic and oncogenic potentials of the interferon regulatory factorss-1 and -2.
Science
259
1993
971
117
Tanaka
N
Ishihara
M
Kitagawa
M
Harada
H
Kimura
T
Matsuyama
T
Lamphier
MS
Aizawa
S
Mak
TW
Taniguchi
T
Cellular commitment to oncogene-induced transformation or apoptosis is dependent on the transcription factor IRF-1.
Cell
77
1994
829
118
Pelicano
L
Li
F
Schnidler
C
Chelbi-Alix
MK
Retinoic acid enhances the expression of interferon-induced proteins: evidence for multiple mechanisms of action.
Oncogene
15
1997
2349
119
Darnell
JE
Jr
STATs and gene regulation.
Science
277
1997
1630
120
Gianni
M
Terao
M
Fortino
I
LiCalzi
M
Viggiano
V
Barbui
T
Rambaldi
A
Garatini
E
Stat1 is induced and activated by all-trans retinoic acid in acute promyelocyitc leukemia cells.
Blood
89
1997
1001
121
Lanotte
M
Martin-Thouvenin
V
Najman
S
Balerini
P
Valensi
F
Berger
R
NB4, a maturation inducible cell line with t(15;17) marker isolated from a human acute promyelocytic leukemia (M3).
Blood
77
1991
1080
122
Chih
DY
Chumakov
AM
Park
DJ
Silla
AG
Koeffler
HP
Modulation of mRNA expression of a novel human myeloid-selective CCAAT/enhancer binding protein gene (C/EBPepsilon).
Blood
90
1997
2987
123
Morosetti
R
Park
DJ
Chumakov
AM
Grillier
I
Shiohara
M
Gombart
AF
Nakamaki
T
Weinberg
K
Koeffler
HP
A novel, myeloid transcription factor, C/EBP epsilon, is upregulated during granulocytic, but not monocytic, differentiation.
Blood
90
1997
2591
124
Yamanaka
R
Kim
GD
Radomska
HS
Lekstrom-Himes
J
Smith
LT
Antonson
P
Tenen
DG
Xanthopoulos
KG
CCAAT/enhancer binding protein epsilon is preferentially up-regulated during granulocytic differentiation and its functional versatility is determined by alternative use of promoters and differential splicing.
Proc Natl Acad Sci USA
94
1997
6462
125
Antonson
P
Stellan
B
Yamanaka
R
Xanthopoulos
KG
A novel human CCAAT/enhancer binding protein gene, C/EBP-epsilon, is expressed in cells of lymphoid and myeloid lineages and is localized on chromosome 14q11.2 close to the T-cell receptor alpha/delta locus.
Genomics
35
1996
30
126
Park
DJ
Chumakov
AM
Grillier
I
Vuong
PT
Silla
AG
Chih
DY
Morosetti
R
Gombart
AF
Koeffler
HP
C/EBPε: The putative target for retinoids in the treatment of acute promyelocytic leukemai.
Blood
90
1997
576a
(abstr, supp 1)
127
Zhu
J
Shi
XG
Chu
HY
Tong
JH
Wang
ZY
Naoe
T
Waxman
S
Chen
SJ
Chen
Z
Effect of retinoic acid isomers on proliferation, differentiation and PML relocalization in the APL cell line NB4.
Leukemia
9
1995
302
128
Drach
J
Lopez-Berestein
G
McQueen
T
Andreeff
M
Mehta
K
Induction of differentiation in myeloid leukemia cell lines and acute promyelocytic leukemia cells by liposomal all-trans-retinoic acid.
Cancer Res
53
1993
2100
129
Brooks
SC
Kazmer
S
Levin
AA
Yen
A
Myeloid differentiation and retinoblastoma phosphorylation changes in HL-60 cells induced by retinoic acid receptor- and retinoid X receptor-selective retinoic acid analogs.
Blood
87
1996
227
130
Liu
M
Iavarone
A
Freedman
LP
Transcriptional activation of the human p21(WAF1/CIP1) gene by retinoic acid receptor. Correlation with retinoid induction of U937 cell differentiation.
J Biol Chem
271
1996
31723
131
Bocchia
M
Xu
Q
Wesley
U
Xu
Y
Koronstsvit
T
Loganzo
F
Albino
AP
Schienberg
DA
Modulation of p53, WAF1/p21 and bcl-2 expression during retinoic acid-induced differentiation of NB4 promnyelocytic cells.
Leuk Res
21
1997
439
132
Wang
ZG
Delva
L
Gaboli
M
Rivi
R
Giorgio
M
Cordon-Cardo
C
Grosveld
F
Pandolfi
PP
Role of PML in cell growth and the retinoic acid pathway.
Science
279
1998
1547
133
Astrom
A
Pettersson
U
Chambon
P
Voorhees
JJ
Retinoic acid induction of human cellular retinoic acid-binding protein-II gene transcription is mediated by retinoic acid receptor-retinoid X receptor heterodimers bound to one far upstream retinoic acid-responsive element with 5-base pair spacing.
J Biol Chem
269
1994
22334
134
Delva
L
Cornic
M
Balitrand
N
Guidez
F
Miclea
JM
Delmer
A
Teillet
F
Fenaux
P
Castaigne
S
Degos
L
Chomienne
C
Resistance to all-trans retinoic acid (ATRA) therapy in relapsing acute promyelocytic leukemia: Study of in vitro ATRA sensitivity and cellular retinoic acid binding protein levels in leukemic cells.
Blood
82
1993
2175
135
Boylan
JF
Gudas
LJ
Overexpression of the cellular retinoic acid binding protein-I (CRABP-I) results in a reduction in differentiation-specific gene expression in F9 teratocarcinoma cells.
J Cell Biol
112
1991
965
136
Mader
S
Thiele
K
Breuer
B
Alonso
A
The promoter of the Hi-zero histone gene contains a DNA element bound by retinoic acid receptors.
J Mol Biol
242
1994
37
137
Nagy
L
Saydak
M
Shipley
N
Lu
S
Basilion
JP
Yan
ZH
Syka
P
Chandraratna
RA
Stein
JP
Heyman
RA
Davies
PJ
Identification and characterization of a versatile retinoid response element (retinoic acid receptor response element-retinoid X receptor response element) in the mouse tissue transglutaminase gene promoter.
J Biol Chem
271
1996
4355
138
Yan
ZH
Noonan
S
Nagy
L
Davies
PJ
Stein
JP
Retinoic acid induction of the tissue transglutaminase promoter is mediated by a novel response element.
Mol Cell Endocrinol
120
1996
203
139
Nakshatri
H
Bouillet
P
Bhat-Nakshatri
P
Chambon
P
Isolation of retinoic acid-repressed genes from P19 embryonal carcinoma cells.
Gene
174
1996
79
140
Bouillet
P
Oulad-Abdelghani
M
Vicaire
S
Garnier
JM
Schuhbaur
B
Dolle
P
Chambon
P
Efficient cloning of cDNAs of retinoic acid-responsive genes in P19 embryonal carcinoma cells and characterization of a novel mouse gene, Stra1 (mouse LERK-2/Eplg2).
Dev Biol
170
1995
420
141
Liu
M
Lee
M
Cohen
M
Bommakanti
M
Freedman
L
Transcriptional activation of the Cdk inhibitor p21 by vitamin D3 leads to the induced differentiation of the myelomonocytic cell line U937.
Genes Dev
10
1996
142
142
Burn
TC
Petrovick
MS
Hohaus
S
Rollins
BJ
Tenen
DG
Monocyte chemoattractant protein-1 gene is expressed in activated neutrophils and retinoic acid-induced human myeloid cell lines.
Blood
84
1994
2776
143
Liang
P
Pardee
AB
Recent advances in differential display.
Cur Opin Immunol
7
1995
274
144
Hubank
M
Schatz
DG
Identifying differences in mRNA expression by representational difference analysis of cDNA.
Nucleic Acids Res
22
1994
5640
145
Yu
M
Tong
JH
Mao
M
Kan
LX
Liu
MM
Sun
YW
Fu
G
Jing
YK
Yu
L
Lepaslier
D
Lanotte
M
Wang
ZY
Chen
Z
Waxman
S
Wang
YX
Tan
JZ
Chen
SJ
Cloning of a gene (RIG-G) associated with retinoic acid-induced differentiation of acute promyelocytic leukemia cells and representing a new member of a family of interferon-stimulated genes.
Proc Natl Acad Sci USA
94
1997
7406
146
Niikura
T
Hirata
R
Weil
SC
A novel interferon-inducible gene expressed during myeloid differentiation.
Blood Cells Mol Dis
23
1997
337
147
Mao
M
Yu
M
Tong
JH
Ye
J
Zhu
J
Huang
QH
Fu
G
Yu
L
Zhao
SY
Waxman
S
Lanotte
M
Wang
ZY
Tan
JZ
Chan
SJ
Chen
Z
RIG-E, a human homolog of the murine Ly-6 family, is induced by retinoic acid during the differentiation of acute promyelocytic leukemia cell.
Proc Natl Acad Sci USA
93
1996
5910
148
Duprez
E
Tong
JH
Derre
J
Chen
SJ
Berger
R
Chen
Z
Lanotte
M
JEM-1, a novel gene encoding a leucine-zipper nuclear factor upregulated during retinoid-induced maturation of NB4 promyelocytic leukaemia.
Oncogene
14
1997
1563
149
Lawson
N
Berliner
N
Calcium/calmodulin dependent protein kinase kinase is an immediate early retinoic acid response gene during neutrophil maturation.
Blood
90
1997
45a
(abstr, supp 1)
150
Scott
L
Mueller
L
Collins
S
E3, a hematopoietic-specific transcript directly regulated by the retinoic receptor α.
Blood
88
1996
2517
151
Adra
C
Zhu
S
Ko
J
Guillemot
J
Cuervo
A
Kobayashi
H
Horiuchi
T
Lelias
J
Rowley
J
Lim
B
LAPTM5: A novel lysosomal-associated multispanning membrane protein preferentially expressed in hematopoietic cells.
Genomics
35
1996
328
152
Schena
M
Shalon
D
Heller
R
Chai
A
Brown
PO
Davis
RW
Parallel human genome analysis: microarray-based expression monitoring of 1000 genes.
Proc Natl Acad Sci USA
93
1996
10614
153
DeRisi
J
Penland
L
Brown
PO
Bittner
ML
Meltzer
PS
Ray
M
Chen
Y
Su
YA
Trent
JM
Use of a cDNA microarray to analyse gene expression patterns in human cancer.
Nat Genet
14
1996
457
154
Lockhart
DJ
Dong
H
Byrne
MC
Follettie
MT
Gallo
MV
Chee
MS
Mittmann
M
Wang
C
Kobayashi
M
Horton
H
Brown
EL
Expression monitoring by hybridization to high density oligonucleotide arrays.
Nat Biotech
14
1996
1675
155
Slack
JL
Ross
DT
Lashkari
D
Shalon
D
Stanley
L
Yu
M
Brown
PO
Use of cDNA microarray technology to analyze gene expression in a myeloid cell line expressing the APL-specific fusion PML-RARα.
Blood
90
1997
321a
(abstr, suppl 1)
156
Tamayo
P
Slonim
D
Mesirou
J
Zhu
Q
Kitareewan
S
Dmitrovsky
E
Lander
ES
Golub
TR
Interpreting patterns of gene expression with self-organizing maps: Methods and application to hematopoietic differentiation.
Proc Natl Acad Sci USA
96
1999
2907
157
de The
H
Lavau
C
Marchio
A
Chomienne
C
Degos
L
Dejean
A
The PML-RAR alpha fusion mRNA generated by the t(15;17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR.
Cell
66
1991
675
158
Stadler
M
Chelbi-Alix
MK
Koken
MH
Venturini
L
Lee
C
Saib
A
Quignon
F
Pelicano
L
Guillemin
MC
Schindler
C
de Thé
H
Transcriptional induction of the PML growth suppressor gene by interferons is mediated through an ISRE and a GAS element.
Oncogene
11
1995
2565
159
Fagioli
M
Alcalay
M
Pandolfi
PP
Venturini
L
Mencarelli
A
Simeone
A
Acampora
D
Grignani
F
Pelicci
PG
Alternative splicing of PML transcripts predicts coexpression of several carboxy-terminally different protein isoforms.
Oncogene
7
1992
1083
160
Saurin
A
Borden
K
Boddy
M
Freemont
P
Does this have a familiar RING?
Trends Biochem Sci
21
1996
208
161
Reddy
B
Etkin
L
A novel zinc finger coiled-coil domain in a family of nuclear proteins.
Trends Biochem Sci
17
1992
344
162
Freemont
PS
The RING finger. A novel protein sequence motif related to the zinc finger.
Ann NY Acad Sci
684
1993
174
163
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/RARalpha protein depend on the fusion of the PML protein dimerization and RARalpha DNA binding domains.
EMBO J
15
1996
4949
164
Borden
KL
Lally
JM
Martin
SR
O’Reilly
NJ
Solomon
E
Freemont
PS
In vivo and in vitro characterization of the B1 and B2 zinc-binding domains from the acute promyelocytic leukemia protooncoprotein PML.
Proc Natl Acad Sci USA
93
1996
1601
165
Borden
K
Freemont
P
The RING finger domain: A recent example of a sequence-structure family.
Curr Opin Struct Biol
6
1996
395
166
Borden
KL
Boddy
MN
Lally
J
O’Reilly
NJ
Martin
S
Howe
K
Solomon
E
Freemont
PS
The solution structure of the RING finger domain from the acute promyelocytic leukaemia proto-oncoprotein PML.
EMBO J
14
1995
1532
167
Le
XF
Yang
P
Chang
KS
Analysis of the growth and transformation suppressor domains of promyelocytic leukemia gene, PML.
J Biol Chem
271
1996
130
168
Borden
KL
Campbell Dwyer
EJ
Salvato
MS
The promyelocytic leukemia protein PML has pro-apoptotic activity mediated through its RING domain.
FEBS Lett
418
1997
30
169
Boddy
MN
Duprez
E
Borden
KL
Freemont
PS
Surface residue mutations of the PML RING finger domain alter the formation of nuclear matrix-associated PML bodies.
J Cell Sci
110
1997
2197
170
Kastner
P
Perez
A
Lutz
Y
Rochette
EC
Gaub
MP
Durand
B
Lanotte
M
Berger
R
Chambon
P
Structure, localization and transcriptional properties of two classes of retinoic acid receptor alpha fusion proteins in acute promyelocytic leukemia (APL): Structural similarities with a new family of oncoproteins.
EMBO J
11
1992
629
171
Perez
A
Kastner
P
Sethi
S
Lutz
Y
Reibel
C
Chambon
P
PMLRAR homodimers: Distinct DNA binding properties and heteromeric interactions with RXR.
EMBO J
12
1993
3171
172
Borden
KL
Campbell Dwyer
EJ
Salvato
MS
An arenavirus RING (zinc-binding) protein binds the oncoprotein promyelocyte leukemia protein (PML) and relocates PML nuclear bodies to the cytoplasm.
J Virol
72
1998
758
173
Daniel
MT
Koken
M
Romagne
O
Barbey
S
Bazarbachi
A
Stadler
M
Guillemin
MC
Degos
L
Chomienne
C
de The
H
PML protein expression in hematopoietic and acute promyelocytic leukemia cells.
Blood
82
1993
1858
174
Chang
KS
Fan
YH
Andreeff
M
Liu
J
Mu
ZM
The PML gene encodes a phosphoprotein associated with the nuclear matrix.
Blood
85
1995
3646
175
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
176
Grotzinger
T
Sternsdorf
T
Jensen
K
Will
H
Interferon-modulated expression of genes encoding the nuclear-dot-associated proteins Sp100 and promyelocytic leukemia protein (PML).
Eur J Biochem
238
1996
554
177
Chelbi-Alix
MK
Pelicano
L
Quignon
F
Koken
MH
Venturini
L
Stadler
M
Pavlovic
J
Degos
L
de The
H
Induction of the PML protein by interferons in normal and APL cells.
Leukemia
9
1995
2027
178
Sternsdorf
T
Jensen
K
Will
H
Evidence for covalent modification of the nuclear dot-associated proteins PML and Sp100 by PIC1/SUMO-1.
J Cell Biol
139
1997
1621
179
Müller
S
Matunis
MJ
Dejean
A
Conjugation with the ubiquitin-related modifier SUMO-1 regulates the partitioning of PML within the nucleus.
EMBO J
17
1998
61
180
Kamitani
T
Kito
K
Nguyen
HP
Wada
H
Fukunda-Kamitani
T
Yeh
EH
Identification of three major sentrinization sites in PML.
J Biol Chem
273
1998
26675
181
Grignani
F
Fagioli
M
Ferrucci
PF
Alcalay
M
Pelicci
PG
The molecular genetics of acute promyelocytic leukemia.
Blood Rev
7
1993
87
182
Mu
ZM
Le
XF
Glassman
AB
Chang
KS
The biologic function of PML and its role in acute promyelocytic leukemia.
Leuk Lymphoma
23
1996
277
183
Terris
B
Baldin
V
Dubois
S
Degott
C
Flejou
JF
Henin
D
Dejean
A
PML nuclear bodies are general targets for inflammation and cell proliferation.
Cancer Res
55
1995
1590
184
Koken
MH
Linares-Cruz
G
Quignon
F
Viron
A
Chelbi-Alix
MK
Sobczak-Thepot
J
Juhlin
L
Degos
L
Calvo
F
de The
H
The PML growth-suppressor has an altered expression in human oncogenesis.
Oncogene
10
1995
1315
185
Gambacorta
M
Flenghi
L
Fagioli
M
Pileri
S
Leoncini
L
Bigerna
B
Pacini
R
Tanci
LN
Pasqualucci
L
Ascani
S
Mencarelli
A
Liso
A
Pelicci
PG
Falini
B
Heterogeneous nuclear expression of the promyelocytic leukemia (PML) protein in normal and neoplastic human tissues.
Am J Pathol
149
1996
2023
186
Aractingi
S
de The
H
Gluckman
E
Le Goue
C
Carosela
ED
PML is expressed in chronic graft-versus-host disease lesions.
Bone Marrow Transplant
19
1997
1125
187
Lam
YW
Ammerlaan
W
O
WS
Kroese
F
Opstelten
D
Cell type-and differentiation stage-dependent expression of PML domains in rat, detected by monoclonal antibody HIS55.
Exp Cell Res
221
1995
344
188
Lavau
C
Dejean
A
The t(15;17) translocation in acute promyelocytic leukemia.
Leukemia
8
1994
S9
189
Nason-Burchenal
K
Gandini
D
Botto
M
Allopenna
J
Seale
JR
Cross
NC
Goldman
JM
Dmitrovsky
E
Pandolfi
PP
Interferon augments PML and PML/RAR alpha expression in normal myeloid and acute promyelocytic cells and cooperates with all-trans retinoic acid to induce maturation of a retinoid-resistant promyelocytic cell line.
Blood
88
1996
3926
190
Wang
Z
Giorgio
M
Rivi
R
Gaboli
M
Pandolfi
P
Growth and tumor suppressive properties of the PML gene of acute promyelocytic leukemia in PML−/− mice.
Blood
88
1996
476a
(abstr, suppl 1)
191
Dyck
JA
Maul
GG
Miller
WH
Jr
Chen
JD
Kakizuka
A
Evans
RM
A novel macromolecular structure is a target of the promyelocyte-retinoic acid receptor oncoprotein.
Cell
76
1994
333
192
Maul
GG
Yu
E
Ishov
AM
Epstein
AL
Nuclear domain 10 (ND10) associated proteins are also present in nuclear bodies and redistribute to hundreds of nuclear sites after stress.
J Cell Biochem
59
1995
498
193
de Thé
G
Riviere
M
Bernhard
W
Examen au microscope electronique de la tumeur VX2 du lapin domestique derivee du papillome de Shope.
Bull Canver
47
1960
569
194
Brasch
K
Ochs
RL
Nuclear bodies (NBS): A newly rediscovered organelle.
Exp Cell Res
202
1992
211
195
Zuchner
D
Sternsdorf
T
Szostecki
C
Heathcote
EJ
Cauch-Dudek
K
Will
H
Prevalence, kinetics, and therapeutic modulation of autoantibodies against Sp100 and promyelocytic leukemia protein in a large cohort of patients with primary biliary cirrhosis.
Hepatology
26
1997
1123
196
Szostecki
C
Guldner
HH
Will
H
Autoantibodies against “nuclear dots” in primary biliary cirrhosis.
Semin Liver Dis
17
1997
71
197
Weis
K
Rambaud
S
Lavau
C
Jansen
J
Carvalho
T
Carmo
FM
Lamond
A
Dejean
A
Retinoic acid regulates aberrant nuclear localization of PML-RAR alpha in acute promyelocytic leukemia cells.
Cell
76
1994
345
198
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
199
Andre
C
Guillemin
MC
Zhu
J
Koken
MH
Quignon
F
Herve
L
Chelbi-Alix
MK
Dhumeaux
D
Wang
ZY
Degos
L
Chen
Z
de The
H
The PML and PML/RARalpha domains: From autoimmunity to molecular oncology and from retinoic acid to arsenic.
Exp Cell Res
229
1996
253
200
Doucas
V
Evans
R
The PML nuclear compartment and cancer.
Biochem Biophys Acta
1288
1996
M25
201
Grande
MA
van der Kraan
I
van Steensel
B
Schul
W
de The
H
van der Voort
HT
de Jong
L
van Driel
R
PML-containing nuclear bodies: Their spatial distribution in relation to other nuclear components.
J Cell Biochem
63
1996
280
202
LaMorte
VJ
Dyck
JA
Ochs
RL
Evans
RM
Localization of nascent RNA and CREB binding protein with the PML-containing nuclear body.
Proc Natl Acad Sci USA
95
1998
4991
203
Vallian
S
Gaken
JA
Gingold
EB
Kouzarides
T
Chang
KS
Farzaneh
F
Modulation of Fos-mediated AP-1 transcription by the promyelocytic leukemia protein.
Oncogene
16
1998
2843
204
Seeler
JS
Marchio
A
Sitterlin
D
Transy
C
Dejean
A
Interaction of SP100 with HP1 proteins: A link between the promyelocytic leukemia-associated nuclear bodies and the chromatin compartment.
Proc Natl Acad Sci USA
95
1998
7316
205
Gall
JG
Tsvetkov
A
Wu
Z
Murphy
C
Is the sphere organelle/coiled body a universal nuclear component?
Dev Genet
16
1995
25
206
Puvion-Dutilleul
F
Chelbi-Alix
M
Koken
M
Quignon
F
Puvion
E
H.
DT
Adenovirus infection induces rearrangements in the intranuclear distribution of the nuclear body-associated PML protein.
Exp Cell Res
218
1995
9
207
Lavau
C
Marchio
A
Fagioli
M
Jansen
J
Falini
B
Lebon
P
Grosveld
F
Pandolfi
PP
Pelicci
PG
Dejean
A
The acute promyelocytic leukaemia-associated PML gene is induced by interferon.
Oncogene
11
1995
871
208
Kamei
H
Reversible large-body formation from nuclear bodies upon amino acid(s) starvation in T24 cells.
Exp Cell Res
224
1996
302
209
Jiang
WQ
Ringertz
N
Altered distribution of the promyelocytic leukemia-associated protein is associated with cellular senescence.
Cell Growth Differ
8
1997
513
210
Mu
ZM
Chin
KV
Liu
JH
Lozano
G
Chang
KS
PML, a growth suppressor disrupted in acute promyelocytic leukemia.
Mol Cell Biol
14
1994
6858
211
Zheng
P
Guo
Y
Niu
Q
Levy
DE
Dyck
JA
Lu
S
Sheiman
LA
Liu
Y
Proto-oncogene PML controls genes devoted to MHC class I antigen presentation.
Nature
396
1998
373
212
Guiochon-Mantel
A
Savouret
J
Quignon
F
Delabre
K
Milgrom
E
de Thé
H
Effect of PML and PML-RAR on the transactivation properties and subcellular localization of steroid hormone receptors.
Mol Endocrinol
9
1995
1791
213
Kuehnle
I
Chu
T
Gruen
J
Margolin
J
The coiled-coil domain of PML is a functionally conserved transcriptional repression domain.
Blood
90
1997
322a
(abstr, suppl 1)
214
Vallian
S
Gäken
JA
Trayner
ID
Gingold
EB
Kourizarides
T
Chang
K-S
Farzaneh
F
Repression of transcription by the promyelocytic leukemia protein, PML.
Exp Cell Res
237
1997
371
215
Vallian
S
Chin
KV
Chang
KS
The promyelocytic leukemia protein interacts with Sp1 and inhibits its transactivation of the epidermal growth factor receptor promoter.
Mol Cell Biol
18
1998
7147
216
Koken
MHM
Reid
A
Quignon
F
Chelbi-Alix
MK
Davies
JM
Kabarowski
JHS
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
217
Grotzinger
T
Jensen
K
Will
H
The interferon (IFN)-stimulated gene Sp100 promoter contains an IFN-gamma activation site and an imperfect IFN-stimulated response element which mediate type I IFN inducibility.
J Biol Chem
271
1996
25253
218
Gongora
C
David
G
Pintard
L
Tissot
C
Hua
TD
Dejean
A
Mechti
N
Molecular cloning of a new interferon-induced PML nuclear body-associated protein.
J Biol Chem
272
1997
19457
219
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
220
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
221
Shen
Z
Pardington-Purtymun
PE
Comeaux
JC
Moyzis
RK
Chen
DJ
UBL1, a human ubiquitin-like protein associating with human RAD51/RAD52 proteins.
Genomics
36
1996
271
222
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
223
Gong
L
Kamitani
T
Fujise
K
Caskey
LS
Yeh
ETH
Preferential Interaction of Sentrin with a Ubiquitin-conjugating Enzyme, Ubc9.
J Biol Chem
272
1997
28198
224
Okura
T
Gong
L
Kamitani
T
Wada
T
Okura
I
Wei
CF
Chang
HM
Yeh
ET
Protection against Fas/APO-1- and tumor necrosis factor-mediated cell death by a novel protein, sentrin.
J Immunol
157
1996
4277
225
Muller
S
Michael
J
Matunis
MJ
Dejean
A
Conjugation with the ubiquitin-related modifier SUMO-1 regulates the partitioning of PML within the nucleus.
EMBO J
17
1998
61
226
Kamitani
T
Kito
K
Nguyen
HP
Wada
H
Fukuda-Kamitani
T
Yeh
ETH
Identification of three major sentrinization sites in PML.
J Biol Chem
273
1998
26675
227
Puccetti
E
Sternsdorf
T
Hoelzer
D
Hans Will
H
Ottmann
OG
Ruthardt
M
PIC-1/SUMO-1 modified PML/RARα is directly involved in arsenic trioxide induced apoptosis in acute promyelocytic leukemia.
Blood
92
1998
212a
(abstr, suppl 1)
228
Ruthardt
M
Orleth
A
Tomassoni
L
Puccetti
E
Riganelli
D
Alcalay
M
Mannucci
R
Nicoletti
I
Grignani
F
Fagioli
M
Pelicci
PG
The acute promyelocytic leukaemia specific PML and PLZF proteins localize to adjacent and functionally distinct nuclear bodies.
Oncogene
16
1998
1945
229
Alcalay
M
Tomassoni
L
Columbo
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
230
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
231
Yacoub
A
Kelley
M
Deutsch
W
Drosophila ribosomal protein P0 contains apurinic/apyrimidinic endonuclease activity.
Nucleic Acid Res
24
1996
4298
232
Uchiumi
T
Ogata
K
Cross-linking study on localization of the binding site for elongation factor 1 alpha on rat liver ribosomes.
J Biol Chem
261
1986
9668
233
Uchiumi
T
Wahba
AJ
Traut
RR
Topography and stoichiometry of acidic proteins in large ribosomal subunits from Artemia salina as determined by crosslinking.
Proc Natl Acad Sci USA
84
1987
5580
234
Redner
RL
Rush
EA
Schlesinger
KW
Pollock
SL
Watkins
SC
The t(5;17) fusion protein NPM-RARα does not alter PML localization.
Blood
90
1997
321a
(abstr, suppl 1)
235
Yung
B
Busch
H
Chan
P
Translocation of nucleolar phosphoprotein B23 (37 kDa/PI 5.1) induced by inhibitors of ribosome synthesis.
Biochim Biophys Acta
826
1985
167
236
Borer
R
Lehner
C
Appenberger
H
Nigg
E
Major nucleolar proteins shuttle between nucleus and cytoplasm.
Cell
56
1989
379
237
Dumbar
T
Gentry
G
Olson
M
Interaction of nuceolar phosphoprotein with nuceic acids.
Biochemistry
28
1989
9495
238
Doucas
V
Ishov
AM
Romo
A
Juguilon
H
Weitzman
MD
Evans
RM
Maul
GG
Adenovirus replication is coupled with the dynamic properties of the PML nuclear structure.
Genes Dev
10
1996
196
239
Everett
R
O’Hare
P
O’Rourke
D
Barlow
P
Orr
A
Point mutations in the herpes simplex virus type 1 Vmw110 RING finger helix affect activation of gene expression, viral growth, and interaction with PML-containing nuclear structures.
J Virol
69
1995
7339
240
Maul
GG
Ishov
AM
Everett
RD
Nuclear domain 10 as preexisting potential replication start sites of herpes simplex virus type-1.
Virology
217
1996
67
241
Jiang
WQ
Szekely
L
Klein
G
Ringertz
N
Intranuclear redistribution of SV40T, p53, and PML in a conditionally SV40T-immortalized cell line.
Exp Cell Res
229
1996
289
242
Szekely
L
Pokrovskaja
K
Jiang
WQ
de The
H
Ringertz
N
Klein
G
The Epstein-Barr virus-encoded nuclear antigen EBNA-5 accumulates in PML-containing bodies.
J Virol
70
1996
2562
243
Korioth
F
Maul
GG
Plachter
B
Stamminger
T
Frey
J
The nuclear domain 10 (ND10) is disrupted by the human cytomegalovirus gene product IE1.
Exp Cell Res
229
1996
155
244
Ahn
J
Hayward
G
The major immediate-early proteins IE1 and IE2 of human cytomegalovirus colocalize with and disrupt PML-associated nuclear bodies at very early times in infected permissive cells.
J Virol
71
1997
4599
245
Day
PM
Roden
RB
Lowy
DR
Schiller
JT
The papillomavirus minor capsid protein, L2, induces localization of the major capsid protein, L1, and the viral transcription/replication protein, E2, to PML oncogenic domains.
J Virol
72
1998
142
246
Chelbi-Alix
MK
Quignon
F
Pelicano
L
Koken
MH
de The
H
Resistance to virus infection conferred by the interferon-induced promyelocytic leukemia protein.
J Virol
72
1998
1043
247
Ahn
MJ
Nason
BK
Moasser
MM
Dmitrovsky
E
Growth suppression of acute promyelocytic leukemia cells having increased expression of the non-rearranged alleles: RAR alpha or PML.
Oncogene
10
1995
2307
248
Liu
JH
Mu
ZM
Chang
KS
PML suppresses oncogenic transformation of NIH/3T3 cells by activated neu.
J Exp Med
181
1995
1965
249
Mu
ZM
Le
XF
Vallian
S
Glassman
AB
Chang
KS
Stable overexpression of PML alters regulation of cell cycle progression in HeLa cells.
Carcinogenesis
18
1997
2063
250
Le
XF
Vallian
S
Mu
ZM
Hung
MC
Chang
KS
Recombinant PML adenovirus suppresses growth and tumorigenicity of human breast cancer cells by inducing G1 cell cycle arrest and apoptosis.
Oncogene
16
1998
1839
251
Fagioli
M
Alcalay
M
Tomassoni
L
Ferrucci
PF
Mencarelli
A
Riganelli
D
Grignani
F
Pozzan
T
Nicoletti
I
Pelicci
PG
Cooperation between the RING + B1-B2 and coiled-coil domains of PML is necessary for its effects on cell survival.
Oncogene
16
1998
2905
252
Quignon
F
De Bels
F
Koken
M
Feuteun
J
Ameisen
J-C
de Thé
H
PML induces a novel caspase-independent death process.
Nat Genet
20
1998
259
253
Wang
Z-G
Ruggero
D
Ronchetti
S
Zhong
S
Gaboli
M
Rivi
R
Pandolfi
PP
PML is essential for multiple apoptotic pathways.
Nat Genet
20
1998
266
254
Hess JL, Korsemeyer SJ: Life, death and nuclear spots. Nat Genet 20, 1998
255
Chan
JY
Li
L
Fan
YH
Mu
ZM
Zhang
WW
Chang
KS
Cell-cycle regulation of DNA damage-induced expression of the suppressor gene PML.
Biochem Biophys Res Commun
240
1997
640
256
Fornace
AJ
Jr
Jackman
J
Hollander
MC
Hoffman-Liebermann
B
Liebermann
DA
Genotoxic-stress-response genes and growth-arrest genes. gadd, MyD, and other genes induced by treatments eliciting growth arrest.
Ann NY Acad Sci
663
1992
139
257
Berghofer-Hochheimer
Y
Zurek
C
Wolfl
S
Hemmerich
P
Munder
T
L7 protein is a coregulator of vitamin D receptor-retinoid X receptor-mediated transactivation.
J Cell Biochem
69
1998
1
258
Jackson
TA
Richer
JK
Bain
DL
Takimoto
GS
Tung
L
Horwitz
KB
The partial agonist activity of antagonist-occupied steroid receptors is controlled by a novel hinge domain-binding coactivator L7/SPA and the corepressors N-CoR or SMRT.
Mol Endocrinol
11
1997
693
259
Wang
Z-G
Rego
E
Peruzzi
D
He
LZ
Pandolfi
PP
Loss of PML function enhances the frequency and onset of APL in PML-RARα transgenic mice.
Blood
92
1998
479a
(abstr, suppl 1)
260
Lo Coco
F
Diverio
D
Pandolfi
PP
Biondi
A
Rossi
V
Avvisati
G
Rambaldi
A
Arcese
W
Petti
MC
Meloni
G
Mandelli
F
Grignani
F
Masera
G
Barbui
T
Pelicci
PG
Molecular evaluation of residual disease as a predictor of relapse in acute promyelocytic leukaemia.
Lancet
340
1992
1437
261
Baranger
L
Gardembas
M
Hillion
J
Foussard
C
Ifrah
N
Boasson
M
Berger
R
Rearrangements of the RARA and PML genes in a cytogenetic variant of acute promyelocytic leukemia.
Genes Chromosomes Cancer
6
1993
118
262
McKinney
CD
Golden
WL
Gemma
NW
Swerdlow
SH
Williams
ME
RARA and PML gene rearrangements in acute promyelocytic leukemia with complex translocations and atypical features.
Genes Chromosomes Cancer
9
1994
49
263
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 leukemia.
Genes Chromosomes Cancer
9
1994
234
264
Hiorns
LR
Min
T
Swansbury
GJ
Zelent
A
Dyer
MJ
Catovsky
D
Interstitial insertion of retinoic acid receptor-alpha gene in acute promyelocytic leukemia with normal chromosomes 15 and 17.
Blood
83
1994
2946
265
Pandolfi
PP
Alcalay
M
Fagioli
M
Zangrilli
D
Mencarelli
A
Diverio
D
Biondi
A
Lo Coco
F
Rambaldi
A
Rochette-Egly
C
Gaube
MP
Chambon
P
Pelicci
PG
Genomic variability and alternative splicing generate multiple PML/RAR alpha transcripts that encode aberrant PML proteins and PML/RAR alpha isoforms in acute promyelocytic leukaemia.
EMBO J
11
1992
1397
266
Dong
S
Geng
JP
Tong
JH
Wu
Y
Cai
JR
Sun
GL
Chen
SR
Wang
ZY
Larsen
CJ
Berger
R
Chen
S-J
Chen
Z
Breakpoint clusters of the PML gene in acute promyelocytic leukemia: Primary structure of the reciprocal products of the PML-RARA gene in a patient with t(15;17).
Genes Chromosomes Cancer
6
1993
133
267
Tong
JH
Dong
S
Geng
JP
Huang
W
Wang
ZY
Sun
GL
Chen
SJ
Chen
Z
Larsen
CJ
Berger
R
Molecular rearrangements of the MYL gene in acute promyelocytic leukemia (APL, M3) define a breakpoint cluster region as well as some molecular variants.
Oncogene
7
1992
311
268
Geng
JP
Tong
JH
Dong
S
Wang
ZY
Chen
SJ
Chen
Z
Zelent
A
Berger
R
Larsen
CJ
Localization of the chromosome 15 breakpoints and expression of multiple PML-RAR alpha transcripts in acute promyelocytic leukemia: A study of 28 Chinese patients.
Leukemia
7
1993
20
269
Chang
KS
Lu
JF
Wang
G
Trujillo
JM
Estey
E
Cork
A
Chu
DT
Freireich
EJ
Stass
SA
The t(15;17) breakpoint in acute promyelocytic leukemia cluster within two different sites of the myl gene: Targets for the detection of minimal residual disease by the polymerase chain reaction.
Blood
79
1992
554
270
Fenaux
P
Chomienne
C
Biology and treatment of acute promyelocytic leukemia.
Curr Opin Oncol
8
1996
3
271
Slack
JL
Arthur
DC
Lawrence
D
Mrozek
K
Mayer
RJ
Davey
FR
Tantravahi
R
Pettenati
MJ
Bigner
S
Carroll
AJ
Rao
KW
Schiffer
CA
Bloomfield
CD
Secondary cytogenetic changes in acute promyelocytic leukemia—Prognostic importance in patients treated with chemotherapy alone and association with the intron 3 breakpoint of the PML gene: A Cancer and leukemia group B study.
J Clin Oncol
15
1997
1786
272
Chen
Z
Chen
A
Tong
JH
Dong
S
Wang
ZY
Waxman
S
Zelent
A
Occurrence of distinct PML-RAR-alpha fusion gene isoforms in patients with acute promyelocytic leukemia detected by reverse transcriptase/polymerase chain reaction.
Oncogene
7
1992
1223
273
Miller
WH
Jr
Kakizuka
A
Frankel
SR
Warrell
RP
Jr
DeBlasio
A
Levine
K
Evans
RM
Dmitrovsky
E
Reverse transcription polymerase chain reaction for the rearranged retinoic acid receptor alpha clarifies diagnosis and detects minimal residual disease in acute promyelocytic leukemia.
Proc Natl Acad Sci USA
89
1992
2694
274
Miller
WH
Jr
Levine
K
DeBlasio
A
Frankel
SR
Dmitrovsky
E
Warrell
RP
Jr
Detection of minimal residual disease in acute promyelocytic leukemia by a reverse transcription polymerase chain reaction assay for the PML/RAR-alpha fusion mRNA.
Blood
82
1993
1689
275
Diverio
D
Rossi
V
Avvisati
G
DeSantis
S
Pistilli
A
Pane
F
Saglio
G
Martinelli
G
Petti
MC
Santoro
A
Pelicci
PG
Mandelli
F
Biondi
A
Coco
FL
Early detection of relapse by prospective reverse transcriptase-polymerase chain reaction analysis of the PML/RARalpha fusion gene in patients with acute promyelocytic leukemia enrolled in the GIMEMA-AIEOP multicenter “AIDA” trial.
Blood
92
1998
784
276
Miller
WH
Jr
Differentiation therapy of acute promyelocytic leukemia: clinical and molecular features.
Cancer Invest
14
1996
142
277
Fenaux
P
Chomienne
C
Biology and treatment of acute promyelocytic leukemia.
Curr Opin Oncol
8
1996
3
278
Huang
W
Sun
GL
Li
XS
Cao
Q
Lu
Y
Jang
GS
Zhang
FQ
Chai
JR
Wang
ZY
Waxman
S
Chen
Z
Chen
S-J
Acute promyelocytic leukemia: clinical relevance of two major PML-RAR alpha isoforms and detection of minimal residual disease by retrotranscriptase/polymerase chain reaction to predict relapse.
Blood
82
1993
1264
279
Vahdat
L
Maslak
P
Miller
WH
Jr
Eardley
A
Heller
G
Scheinberg
DA
Warrell
RP
Jr
Early mortality and the retinoic acid syndrome in acute promyelocytic leukemia: Impact of leukocytosis, low-dose chemotherapy, PML/RAR-alpha isoform, and CD13 expression in patients treated with all-trans retinoic acid.
Blood
84
1994
3843
280
Gallagher
RE
Li
YP
Rao
S
Paietta
E
Andersen
J
Etkind
P
Bennett
JM
Tallman
MS
Wiernik
PH
Characterization of acute promyelocytic leukemia cases with PML-RAR alpha break/fusion sites in PML exon 6: Identification of a subgroup with decreased in vitro responsiveness to all-trans retinoic acid.
Blood
86
1995
1540
281
Al-Omar
H
Khalil
S
Sher
G
Kamel-Reid
S
The short form of PML-RARA gene fusion in acute promyelocytic leukemia is associated morphologically with the presence of primitive myeloblasts.
Blood
90
1997
207b
(abstr, suppl 1)
282
Fukutani
H
Naoe
T
Ohno
R
Yoshida
H
Miyawaki
S
Shimazaki
C
Miyake
T
Nakayama
Y
Kobayashi
H
Goto
S
Takeshita
A
Kobayashi
S
Kato
Y
Shiraishi
K
Sasada
M
Ohtake
S
Murakami
H
Kobayashi
M
Endo
N
Shindo
H
Matsushita
K
Hasegawa
S
Tsuji
K
Ueda
Y
Tominaga
N
Furuya
H
Inoue
Y
Takeuchi
J
Morishita
H
Iida
H
Isoforms of PML-retinoic acid receptor alpha fused transcripts affect neither clinical features of acute promyelocytic leukemia nor prognosis after treatment with all-trans retinoic acid. The Leukemia Study Group of the Ministry of Health and Welfare (Kohseisho).
Leukemia
9
1995
1478
283
Fukutani
H
Naoe
T
Yoshida
H
Yamamori
S
Ohno
R
Molecular heterogeneity of the PML gene rearrangement in acute promyelocytic leukemia: Prevalence and clinical significance.
Jpn J Cancer Res
84
1993
257
284
Dong
S
Tong
JH
Huang
W
Chen
SJ
Chen
Z
Wang
ZY
Geng
JP
Qi
ZW
Molecular study on the chromosome 15 breakpoints in the translocation t(15; 17) in acute promyelocytic leukemia (APL).
Sci China B Chem Life Sci Earth Sci
36
1993
1101
285
Tashiro
S
Kotomura
N
Tanaka
K
Suzuki
K
Kyo
T
Dohy
H
Niwa
O
Kamada
N
Identification of illegitimate recombination hot spot of the retinoic acid receptor alpha gene involved in 15;17 chromosomal translocation of acute promyelocytic leukemia.
Oncogene
9
1994
1939
286
Yoshida
H
Naoe
T
Fukutani
H
Kiyoi
H
Kubo
K
Ohno
R
Analysis of the joining sequences of the t(15;17) translocation in human acute promyelocytic leukemia: sequence non-specific recombination between the PML and RARA genes within identical short stretches.
Genes Chromosomes Cancer
12
1995
37
287
Jansen
JH
Mahfoudi
A
Rambaud
S
Lavau
C
Wahli
W
Dejean
A
Multimeric complexes of the PML-retinoic acid receptor alpha fusion protein in acute promyelocytic leukemia cells and interference with retinoid and peroxisome-proliferator signaling pathways.
Proc Natl Acad Sci USA
92
1995
7401
288
Leid
M
Kastner
P
Lyons
R
Nakshatri
H
Saunders
M
Zacharewski
T
Chen
JY
Staub
A
Garnier
JM
Mader
S
Chambon
P
Purification, cloning, and RXR identity of the HeLa cell factor with which RAR or TR heterodimerizes to bind target sequences efficiently.
Cell
68
1992
377
289
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
290
Nervi
C
Poindexter
EC
Grignani
F
Pandolfi
PP
Lo Coco
F
Avvisati
G
Pelicci
PG
Jetten
AM
Characterization of the PML-RAR alpha chimeric product of the acute promyelocytic leukemia-specific t(15;17) translocation.
Cancer Res
52
1992
3687
291
Benedetti
L
Levin
AA
Scicchitano
BM
Grignani
F
Allenby
G
Diverio
D
Lo Coco
F
Avvisati
G
Ruthardt
M
Adamo
S
Pelicci
PG
Nervi
C
Characterization of the retinoid binding properties of the major fusion products present in acute promyelocytic leukemia cells.
Blood
90
1997
1175
292
Rousselot
P
Hardas
B
Patel
A
Guidez
F
Gaken
J
Castaigne
S
Dejean
A
de The
H
Degos
L
Farzaneh
F
Chomienne
C
The PML-RAR alpha gene product of the t(15;17) translocation inhibits retinoic acid-induced granulocytic differentiation and mediated transactivation in human myeloid cells.
Oncogene
9
1994
545
293
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
294
Guidez
F
Ivins
S
Zhu
J
Söderström
M
Waxman
S
Zelent
A
Reduced retinoic acid-sensitivities of nuclear receptor co-repressor binding to PML- and PLZF-RARa underlie molecular pathogenesis and treatment of acute promyelocytic leukemia.
Blood
91
1998
2634
295
Lin
RJ
Nagy
L
Inoue
S
Shao
W
Miller
WH
Jr
Evans
RM
Role of the histone deacetylase complex in acute promyelocytic leukemia.
Nature
391
1998
811
296
Grignani
F
De Metteis
S
Nervi
C
Tomassoni
L
Gelmetti
V
Cioce
M
Fanelli
M
Ruthardt
M
Ferrara
FF
Zamir
I
Seiser
C
Grignani
F
Lazar
MA
Minucci
S
Pelicci
PG
Fusion proteins of the retinoic acid receptor-a recruit histone deacteylase in promyelocytic leukemia.
Nature
391
1998
815
297
Candido
EP
Reeves
R
Davie
JR
Sodium butyrate inhibits histone deacetylation in cultured cells.
Cell
14
1978
105
298
Yoshida
M
Horinouchi
S
Beppu
T
Trichostatin A and trapoxin: Novel chemical probes for the role of histone acetylation in chromatin structure and function.
Bioessays
17
1995
423
299
He
L-Z
Guidez
F
Triboli
C
Peruzzi
D
Ruthardt
M
Zelent
A
Pandolfi
PP
Distinct interactions of PML-RARα and PLZF-RARα with co-repressors determine differential responses to RA in APL.
Nat Gen
18
1998
126
300
Chen
A
Licht
JD
Wu
Y
Hellinger
N
Scher
W
Waxman
S
Retinoic acid is required for and potentiates differentiation of acute promyelocytic leukemia cells by nonretinoid agents.
Blood
84
1994
2122
301
Doucas
V
Brockes
JP
Yaniv
M
de The
H
Dejean
A
The PML-retinoic acid receptor alpha translocation converts the receptor from an inhibitor to a retinoic acid-dependent activator of transcription factor AP-1.
Proc Natl Acad Sci USA
90
1993
9345
302
Dermime
S
Grignani
F
Clerici
M
Nervi
C
Sozzi
G
Talamo
GP
Marchesi
E
Formelli
F
Parmiani
G
Pelicci
PG
Gambacorti-Passerini
C
Occurrence of resistance to retinoic acid in the acute promyelocytic leukemia cell line NB4 is associated with altered expression of the pml/RAR alpha protein.
Blood
82
1993
1573
303
Fanelli
M
Nervi
C
Pelicci
PG
Gambacorti-Passerini
C
Constitutive degradation of the PML/RARa protein is present in retinoic acid (RA)-resistant acute promyelocytic leukemia cells and involves the proteosome.
Blood
90
1997
70a
(abstr, suppl 1)
304
Duprez
E
Ruchaud
S
Houge
G
Martin-Thouvenin
V
Valensi
F
Kastner
P
Berger
R
Lanotte
M
A retinoid acid ‘resistant’ t(15;17) acute promyelocytic leukemia cell line: Isolation, morphological, immunological, and molecular features.
Leukemia
6
1992
1281
305
Rosenauer
A
Raelson
JV
Nervi
C
Eydoux
P
DeBlasio
A
Miller
WH
Jr
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
306
Shao
W
Benedetti
L
Lamph
WW
Nervi
C
Miller
WH
Jr
A retinoid-resistant acute promyelocytic leukemia subclone expresses a dominant negative PML-RAR alpha mutation.
Blood
89
1997
4282
307
Imaizumi
M
Suzuki
H
Yoshinari
M
Sato
A
Saito
T
Sugawara
A
Tsuchiya
S
Hatae
Y
Fujimoto
T
Kakizuka
A
Konno
T
Iinuma
K
Mutations in the E-domain of RAR portion of the PML/RAR chimeric gene may confer clinical resistance to all-trans retinoic acid in acute promyelocytic leukemia.
Blood
92
1998
374
308
Ding
W
Li
YP
Nobile
LM
Grills
G
Carrera
I
Paietta
E
Tallman
MS
Wiernik
PH
Gallagher
RE
Leukemic cellular retinoic acid resistance and missense mutations in the PML-RARalpha fusion gene after relapse of acute promyelocytic leukemia from treatment with All-trans retinoic acid and intensive chemotherapy.
Blood
92
1998
1172
309
Kizaki
M
Matsushita
H
Takayama
N
Muto
A
Ueno
H
Awaya
N
Kawai
Y
Asou
H
Kamada
N
Ikeda
Y
Establishment and characterization of a novel acute promyelocytic leukemia cell line (UF-1) with retinoic acid-resistant features.
Blood
88
1996
1824
310
Duprez
E
Lillehaug
JR
Naoe
T
Lanotte
M
cAMP signalling is decisive for recovery of nuclear bodies (PODs) during maturation of RA-resistant t(15;17) promyelocytic leukemia NB4 cells expressing PML-RAR alpha.
Oncogene
12
1996
2451
311
Zhu
J
Koken
M
Quignon
F
Chelbi-Alix
M
Degos
L
Wang
Z
Chen
Z
de The
H
Arsenic-induced PML targeting onto nuclear bodies: Implications for the treatment of acute promyelocytic leukemia.
Proc Natl Acad Sci USA
94
1997
3978
312
Duprez
E
Lillehaug
JR
Gaub
MP
Lanotte
M
Differential changes of retinoid-X-receptor (RXR alpha) and its RAR alpha and PML-RAR alpha partners induced by retinoic acid and cAMP distinguish maturation sensitive and resistant t(15;17) promyelocytic leukemia NB4 cells.
Oncogene
12
1996
2443
313
Nervi
C
Ferrara
FF
Fanelli
M
Rippo
MR
Tomassini
B
Ferrucci
PF
Ruthardt
M
Gelmetti
V
Gambacorti-Passerini
C
Diverio
D
Grignani
F
Pelicci
PG
Testi
R
Caspases mediate retinoic acid-induced degradation of the acute promyelocytic leukemia PML/RARalpha fusion protein.
Blood
92
1998
2244
314
Slack
JL
Yu
M
Constitutive expression of the promyelocytic leukemia-associated oncogene PML-RARalpha in TF1 cells: Isoform-specific and retinoic acid-dependent effects on growth, bcl-2 expression, and apoptosis.
Blood
91
1998
3347
315
Shen
ZX
Chen
GQ
Ni
JH
Li
XS
Xiong
SM
Qiu
QY
Zhu
J
Tang
W
Sun
GL
Yang
KQ
Chen
Y
Zhou
L
Fang
ZW
Wang
YT
Ma
J
Zhang
P
Zhang
TD
Chen
SJ
Chen
Z
Wang
ZY
Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients.
Blood
89
1997
3354
316
Chen
GQ
Zhu
J
Shi
XG
Ni
JH
Zhong
HJ
Si
GY
Jin
XL
Tang
W
Li
XS
Xong
SM
Shen
ZX
Sun
GL
Ma
J
Zhang
P
Zhang
TD
Gazin
C
Naoe
T
Chen
SJ
Wang
ZY
Chen
Z
In vitro studies on cellular and molecular mechanisms of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia: As2O3 induces NB4 cell apoptosis with downregulation of Bcl-2 expression and modulation of PML-RAR alpha/PML proteins.
Blood
88
1996
1052
317
Andre
C
Guillemin
MC
Zhu
J
Koken
MH
Quignon
F
Herve
L
Chelbi-Alix
MK
Dhumeaux
D
Wang
ZY
Degos
L
Chen
Z
de Thé
H
The PML and PML/RARalpha domains: from autoimmunity to molecular oncology and from retinoic acid to arsenic.
Exp Cell Res
229
1996
253
318
Shao
W
Fanelli
M
Ferrara
FF
Riccioni
R
Rosenauer
A
Davison
K
Lamph
WW
Waxman
S
Pelicci
PG
Lo Coco
F
Avvisati
G
Testa
U
Peschle
C
Gambacorti-Passerini
C
Nervi
C
Miller
WH
Jr
Arsenic trioxide as an inducer of apoptosis and loss of PML/RAR alpha protein in acute promyelocytic leukemia cells.
J Natl Cancer Inst
90
1998
124
319
Gianni
M
Koken
MH
Chelbi-Alix
MK
Benoit
G
Lanotte
M
Chen
Z
de The
H
Combined arsenic and retinoic acid treatment enhances differentiation and apoptosis in arsenic-resistant NB4 cells.
Blood
91
1998
4300
320
Chen
GQ
Shi
XG
Tang
W
Xiong
SM
Zhu
J
Cai
X
Han
ZG
Ni
JH
Shi
GY
Jia
PM
Liu
MM
He
KL
Niu
C
Ma
J
Zhang
P
Zhang
TD
Paul
P
Naoe
T
Kitamura
K
Miller
W
Waxman
S
Wang
ZY
de The
H
Chen
SJ
Chen
Z
Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): I. As2O3 exerts dose-dependent dual effects on APL cells.
Blood
89
1997
3345
321
Wang
ZG
Rivi
R
Delva
L
Konig
A
Scheinberg
DA
Gambacorti-Passerini
C
Gabrilove
JL
Warrell
RP
Pandolfi
PP
Arsenic trioxide and melarsoprol induce programmed cell death in myeloid leukemia cell lines and function in a PML and PML-RARalpha independent manner.
Blood
92
1998
1497
322
Konig
A
Wrazel
L
Warrell
RP
Jr
Rivi
R
Pandolfi
PP
Jakubowski
A
Gabrilove
JL
Comparative activity of melarsoprol and arsenic trioxide in chronic B-cell leukemia lines.
Blood
90
1997
562
323
Müller
S
Miller
WH
Dejean
A
Trivalent antimonials induce degradation of the PML-RAR oncoprotein and reorganization of the promyelocytic leukemia nuclear bodies in acute promyelocytic leukemia NB4 cells.
Blood
92
1998
4308
324
Grignani
F
Ferrucci
PF
Testa
U
Talamo
G
Fagioli
M
Alcalay
M
Mencarelli
A
Grignani
F
Peschle
C
Nicoletti
I
Pelicci
PG
The acute promyelocytic leukemia-specific PML-RAR alpha fusion protein inhibits differentiation and promotes survival of myeloid precursor cells.
Cell
74
1993
423
325
Lavau
C
Heard
JM
Danos
O
Dejean
A
Retroviral vectors for the transduction of the PML-RARalpha fusion product of acute promyelocytic leukemia.
Exp Hematol
24
1996
544
326
Ferrucci
PF
Grignani
F
Pearson
M
Fagioli
M
Nicoletti
I
Pelicci
PG
Cell death induction by the acute promyelocytic leukemia-specific PML/RARalpha fusion protein.
Proc Natl Acad Sci USA
94
1997
10901
327
Testa
U
Grignani
F
Barberi
T
Fagioli
M
Masciulli
R
Ferrucci
PF
Seripa
D
Camagna
A
Alcalay
M
Pelicci
PG
Peschle
C
PML/RAR alpha+ U937 mutant and NB4 cell lines: retinoic acid restores the monocytic differentiation response to vitamin D3.
Cancer Res
54
1994
4508
328
Testa
U
Grignani
F
Samoggia
P
Zanetti
C
Riccioni
R
Coco
FL
Diverio
D
Felli
N
Passerini
CG
Grell
M
Pelicci
PG
Peschle
C
The PML/RARalpha fusion protein inhibits tumor necrosis factor-alpha-induced apoptosis in U937 cells and acute promyelocytic leukemia blasts.
J Clin Invest
101
1998
2278
329
Dubois
C
Schlageter
MH
de Gentile
A
Guidez
F
Balitrand
N
Toubert
ME
Krawice
I
Fenaux
P
Castaigne
S
Najean
Y
Hematopoietic growth factor expression and ATRA sensitivity in acute promyelocytic blast cells.
Blood
83
1994
3264
330
Rogaia
D
Grignani
F
Grignani
F
Nicoletti
I
Pelicci
PG
The acute promyelocytic leukemia-specific PML/RAR alpha fusion protein reduces the frequency of commitment to apoptosis upon growth factor deprivation of GM-CSF-dependent myeloid cells.
Leukemia
9
1995
1467
331
Nason-Burchenal
K
Allopenna
J
Begue
A
Stehelin
D
Dmitrovsky
E
Martin
P
Targeting of PML/RARalpha is lethal to retinoic acid-resistant promyelocytic leukemia cells.
Blood
92
1998
1758
332
Nason-Burchenal
K
Takle
G
Pace
U
Flynn
S
Allopenna
J
Martin
P
George
ST
Goldberg
AR
Dmitrovsky
E
Targeting the PML/RAR alpha translocation product triggers apoptosis in promyelocytic leukemia cells.
Oncogene
17
1998
1759
333
Kitamura
K
Yoshida
H
Ohno
R
Naoe
T
Toxic effects of arsenic (As3+) and other metal ions on acute promyelocytic leukemia cells.
Int J Hematol
65
1997
179
185
334
Dai
J
Weinberg
RS
Waxman
S
Jing
Y
Malignant cells can be sensitized to undergo growth inhibition and apoptosis by As2O3 through modification of the glutathione redox system.
Blood
93
1999
268
335
Polyak
K
Xia
Y
Zweier
JL
Kinzler
KW
Vogelstein
B
A model for p53-induced apoptosis.
Nature
389
1997
300
336
Lavau
C
Du
C
Redner
R
Dejean
A
Transformation of primary murine hematopoietic cells by retroviral transduction with wild-type or fusion mutants of the retinoic acid receptor.
Blood
90
1997
320a
(abstr, suppl 1)
337
Altabef
M
Garcia
M
Lavau
C
Bae
S-C
Dejean
A
Samarut
J
A retrovirus carrying the promyelocyte-retinoic acid receptor PML-RARα fusion gene transforms haematopoietic progenitors in vitro and induces acute leukemia.
EMBO J
15
1996
2707
338
Tsai
S
Bartelmez
S
Sitnicka
E
Collins
S
Lymphohematopoietic progenitors immortalized by a retroviral vector harboring a dominant-negative retinoic acid receptor can recapitulate lymphoid, myeloid, and erythroid development.
Genes Dev
8
1994
2831
339
Early
E
Moores
M
Kakizuka
A
Nason-Burchenal
K
Martin
P
Evans
R
Dmitrowsky
Transgenic expression of PML-RARα impairs myelopoiesis.
Proc Natl Acad Sci USA
93
1996
7900
340
David
G
Terris
B
Marchio
A
Lavau
C
Dejean
A
The acute promyelocytic leukemia PML-RAR alpha protein induces hepatic preneoplastic and neoplastic lesions in transgenic mice.
Oncogene
14
1997
1547
341
Grisolano
J
Wesselschmidt
R
Pellici
P
Ley
T
Altered myeloid development and acute leukemia in transgenic mice expressing PML-RARα under control of cathepsin G regulatory sequences.
Blood
89
1997
376
342
He
LZ
Tribioli
C
Rivi
R
Peruzzi
D
Pelicci
PG
Soares
V
Cattoretti
G
Pandolfi
PP
Acute leukemia with promyelocytic features in PML/RARalpha transgenic mice.
Proc Natl Acad Sci USA
94
1997
5302
343
Lagasse
E
Weissman
IL
Mouse MRP8 and MRP14, two intracellular calcium-binding proteins associated with the development of the myeloid lineage.
Blood
79
1992
1907
344
Hanson
RD
Connolly
NL
Burnett
D
Campbell
EJ
Senior
RM
Ley
TJ
Developmental regulation of the human cathepsin G gene in myelomonocytic cells.
J Biol Chem
265
1990
1524
345
Brown
D
Kogan
S
Lagasse
E
Weissman
I
Alcalay
M
Pelicci
PG
Atwater
S
Bishop
JM
A PMLRARalpha transgene initiates murine acute promyelocytic leukemia.
Proc Natl Acad Sci USA
94
1997
2551
346
Turhan
AG
Lemoine
FM
Debert
C
Bonnet
ML
Baillou
C
Picard
F
Macintyre
EA
Varet
B
Highly purified primitive hematopoietic stem cells are PML-RARA negative and generate nonclonal progenitors in acute promyelocytic leukemia.
Blood
85
1995
2154
347
Corral
J
Lavenir
I
Impey
H
Warren
A
Forster
A
Larson
T
Bell
S
McKenzie
A
King
G
Rabbitts
T
An Mll-AF9 fusion gene made by homologous recombination cause acute promyelocytic leukemia in chimeric mice: A method to create fusion oncogenes.
Cell
85
1996
853
348
MacKenzie
KL
Dmitrovsky
E
Moore
MAS
PML-RARα and N-ras synergize to promote proliferation of myelomonocytic cells.
Blood
90
1997
323a
(abstr, suppl 1)
349
Ruthardt
M
Testa
U
Nervi
C
Ferrucci
PF
Grignani
F
Puccetti
E
Grignani
F
Peschle
C
Pelicci
PG
Opposite effects of the acute promyelocytic leukemia PML-retinoic acid receptor alpha (RAR alpha) and PLZF-RAR alpha fusion proteins on retinoic acid signalling.
Mol Cell Biol
17
1997
4859
350
Ruchaud
S
Duprez
E
Gendron
MC
Houge
G
Genieser
HG
Jastorff
B
Doskeland
SO
Lanotte
M
Two distinctly regulated events, priming and triggering, during retinoid-induced maturation and resistance of NB4 promyelocytic leukemia cell line.
Proc Natl Acad Sci USA
91
1994
8428
351
Alcalay
M
Zangrilli
D
Fagioli
M
Pandolfi
PP
Mencarelli
A
Lo Coco
F
Biondi
A
Grignani
F
Pelicci
PG
Expression pattern of the RAR alpha-PML fusion gene in acute promyelocytic leukemia.
Proc Natl Acad Sci USA
89
1992
4840
352
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. M.R.C. Adult Leukaemia Working Party.
Br J Haematol
94
1996
557
353
Li
YP
Andersen
J
Zelent
A
Rao
S
Paietta
E
Tallman
MS
Wiernik
PH
Gallagher
RE
RAR alpha1/RAR alpha2-PML mRNA expression in acute promyelocytic leukemia cells: A molecular and laboratory-clinical correlative study.
Blood
90
1997
306
354
Tobal
K
Saunders
MJ
Grey
MR
Yin
JA
Persistence of RAR alpha-PML fusion mRNA detected by reverse transcriptase polymerase chain reaction in patients in long-term remission of acute promyelocytic leukaemia.
Br J Haematol
90
1995
615
355
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
356
Pollock
J
Grisolano
J
Goda
P
Westervelt
P
Ley
T
Development of acute promyelocytic leukemia (APML) in transgenic mice expressing both the PML-RARα and the reciprocal fusion protein, RARα-PML.
Blood
90
1997
320a
(abstr, suppl 1)
357
Chen
ZX
Xue
Y
Zhang
R
Tao
R
Xia
X
Li
C
Wang
W
Zu
W
Yao
X
Ling
B
A clinical and experimental study on all-transretinoic acid-treated acute promyelocytic leukemia patients.
Blood
78
1991
1413
358
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
359
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
360
Guidez
F
Huang
W
Tong
J-T
Dubois
C
Balitrand
N
Michaux
J
Martiat
P
Degos
L
Chen
Z
Chomienne
C
Poor response to all-trans retinoic acid in a t(11;17) PLZF/RARα AML3 patient.
Leukemia
8
1994
312
361
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
362
Grimwade
D
Gorman
P
Duprez
E
Howe
K
Langabeer
S
Oliver
F
Walker
H
Culligan
D
Waters
J
Pomfret
M
Goldstone
A
Burnett
A
Freemont
P
Sheer
D
Solomon
E
Characterization of cryptic rearrangements and variant translocations in acute promyelocytic leukemia.
Blood
90
1997
4876
363
Culligan
DJ
Stevenson
D
Chee
YL
Grimwade
D
Acute promyelocytic leukaemia with t(11;17)(q23;q12-21) and a good initial response to prolonged ATRA and combination chemotherapy.
Br J Haematol
100
1998
328
364
Grimwade
D
Biondi
A
Mozziconacci
M-J
Hagemeijer
A
Berger
R
Neat
M
Howe
K
Jansen
J
Radford-Weiss
I
Head
D
Liso
V
Sainty
D
Flandrin
G
Birg
F
Lafage-Pochitaloff
M
Characterisation of acute promyelocytic leukaemia (apl) cases lacking the classical t(15;17): Results of the European Working Party.
Blood
92
1998
677a
(abstr, suppl 1)
365
Jansen
JH
de Ridder
MC
Erpelinck
C
Geertsma
W
de Greef
GE
Löwenberg
B
Granulocytic differentiation of t(11;17) positive AML-M3 cells.
Blood
88
1996
173b
(abstr, suppl 1)
366
Jansen
JH
de Ridder
MC
Geertsma
WMC
Erpelinck
CAJ
Smit
B
Slater
R
vd Reijden
BA
de Greef
GE
Sonneveld
P
Löwenberg
B
Complete remission of t(11;17) positive acute promyelocytic leukemia by all-trans retinoic acid and G-CSF.
Blood
92
1998
402a
(abstr, suppl 1)
367
Cook
M
Gould
A
Brand
N
Davies
J
Strutt
P
Shaknovich
R
Licht
J
Waxman
S
Chen
Z
Gluecksohn-Waelsch
S
Krumlauf
R
Zelent
A
Expression of the zinc-finger gene PLZF at rhombomere boundaries in the vertebrate hindbrain.
Proc Natl Acad Sci USA
92
1995
2249
368
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
369
Downing
JR
Look
AT
MLL fusion genes in the 11q23 acute leukemias.
Cancer Treat Res
84
1996
73
370
Rowley
JD
Rearrangements involving chromosome band 11Q23 in acute leukaemia.
Semin Cancer Biol
4
1993
377
371
372
Baysal
BE
van Schothorst
EM
Farr
JE
James
MR
Devilee
P
Richard
CWr
A high-resolution STS, EST, and gene-based physical map of the hereditary paraganglioma region on chromosome 11q23.
Genomics
44
1997
214
373
Bardwell
VJ
Treisman
R
The POZ domain: A conserved protein-protein interaction motif.
Genes Dev
8
1994
1664
374
Chang
CC
Ye
BH
Chaganti
RS
Dalla-Favera
R
BCL-6, a POZ/zinc-finger protein, is a sequence-specific transcriptional repressor.
Proc Natl Acad Sci USA
93
1996
6947
375
Albagli
O
Dhordain
P
Bernadin
F
Quief
S
Kerkaert
J
Leprince
D
Multiple domains participate in distance independent LAZ3/Bcl6 mediated transcriptional repression.
Biophys Biochem Res Commun
220
1996
911
376
Seyfert
VL
Allman
D
He
Y
Staudt
LM
Transcriptional repression by the proto-oncogene BCL-6.
Oncogene
12
1996
2331
377
Numoto
M
Niwa
O
Kaplan
J
Wong
KK
Merrell
K
Kamiya
K
Yanagihara
K
Calame
K
Transcriptional repressor ZF5 identifies a new conserved domain in zinc finger proteins.
Nucleic Acids Res
21
1993
3767
378
Oyake
T
Itoh
K
Motohashi
H
Hayashi
N
Hoshino
H
Nishizawa
M
Yamamoto
M
Igarashi
K
Bach proteins belong to a novel family of BTB-basic leucine zipper transcription factors that interact with MafK and regulate transcription through the NF-E2 site.
Mol Cell Biol
16
1996
6083
379
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 Diff
6
1995
1193
380
Raff
J
Kellum
R
Alberts
B
The Drosophila GAGA transcription factor is associated with specific regions of heterochromatin throughout the cell cycle.
EMBO J
13
1995
5977
381
Li
X
Lopez-Guisa
JM
Ninan
N
Weiner
EJ
Rauscher
FJ
III
Marmorstein
R
Overexpression, purification, characterization, and crystallization of the BTB/POZ domain from the PLZF oncoprotein.
J Biol Chem
272
1997
27324
382
Ahmad
KF
Engel
CK
Prive
GG
Crystal structure of the BTB domain from PLZF.
Proc Natl Acad Sci USA
95
1998
12123
383
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 the activites of promyelocytic leukemia associated PLZF-RARα fusion protein.
Proc Natl Acad Sci USA
93
1996
3624
384
Hummel
J
Wells
R
Dubé
I
Licht
J
Kamel-Reid
S
Deregulation of NPM and PLZF in a variant t(5;17) case of acute promyelocytic leukemia.
Oncogene
18
1999
633
385
Parrado
A
Padua
R
McKenna
S
Dupas
S
Davis
J
Zelent
A
Legal
I
Bentley
P
Whitaker
J
Sigaux
F
Chomienne
C
Abnormal promyelocyric leukemia zinc finger (PLZF) expression in B-CLL.
Blood
88
1996
110b
(abstr, suppl 1)
386
Ivins
S
Zelent
A
Promyelocytic leukaemia translocation gene, PLZF, encodes a transcriptional repressor targeting HOXB2 expression.
Blood
92
1998
308a
(abstr, suppl 1)
387
Avantaggiato
V
Pandolfi
P
Ruthardt
M
Hawe
N
Acampora
D
Pelicci
P
Simeone
A
Developmental analysis of murine promyelocyte leukemia zinc finger (PLZF) gene expression: Implications for the neuromeric model of the forebrain organization.
J Neurosci
15
1995
4927
388
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
291a
(abstr, supp 1)
389
Sassone-Corsi
P
Transcriptional checkpoints determining the fate of male germ cells.
Cell
88
1997
163
390
Toscani
A
Mettus
RV
Coupland
R
Simpkins
H
Litvin
J
Orth
J
Hatton
KS
Reddy
EP
Arrest of spermatogenesis and defective breast development in mice lacking A-myb.
Nature
386
1997
713
391
Sweeney
C
Murphy
M
Kubelka
M
Ravnik
SE
Hawkins
CF
Wolgemuth
DJ
Carrington
M
A distinct cyclin A is expressed in germ cells in the mouse.
Development
122
1996
53
392
Ravnik
SE
Wolgemuth
DJ
The developmentally restricted pattern of expression in the male germ line of a murine cyclin A, cyclin A2, suggests roles in both mitotic and meiotic cell cycles.
Dev Biol
173
1996
69
393
Li
J-Y
English
MA
Bischt
S
Waxman
S
Licht
JD
DNA binding and trancriptional regulation by the promyelocytic leukemia zinc finger protein.
Blood
84
1994
41a
(abstr, suppl 1)
394
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
395
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
396
Pavletich
NP
Pabo
CO
Crystal structure of a five-finger GLI-DNA complex: New perspectives on zinc fingers.
Science
261
1993
1701
397
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
398
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
399
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
400
Dhordain
P
Albagli
O
Lin
RJ
Ansieau
S
Quief
S
Leutz
A
Kerckaert
JP
Evans
RM
Leprince
D
Corepressor SMRT binds the BTB/POZ repressing domain of the LAZ3/BCL6 oncoprotein.
Proc Natl Acad Sci USA
94
1997
10762
401
Ball
HJ
Zelent
A
Licht
JD
The promyelocytic leukemia zinc finger protein binds to DNA in complex containing a cyclin dependent kinase.
Blood
92
1998
213a
(abstr, suppl 1)
402
Long
JJ
Leresche
A
Kriwacki
RW
Gottesfeld
JM
Repression of TFIIH transcriptional activity and TFIIH-associated cdk7 kinase activity at mitosis.
Mol Cell Biol
18
1998
1467
403
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
404
Sherr
CJ
G1 phase progression: Cycling on cue.
Cell
79
1994
551
405
Sherr
CJ
Cancer cell cycles.
Science
274
1996
1672
406
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
407
Yang
E
Korsmeyer
SJ
Molecular thanatopsis: A discourse on the BCL2 family and cell death.
Blood
88
1996
386
408
Chen
Z
Guidez
F
Rousselot
P
Agadir
A
Chen
SJ
Wang
ZY
Degos
L
Zelent
A
Waxman
S
Chomienne
C
PLZF-RAR alpha fusion proteins generated from the variant t(11;17)(q23;q21) translocation in acute promyelocytic leukemia inhibit ligand-dependent transactivation of wild-type retinoic acid receptors.
Proc Natl Acad Sci USA
91
1994
1178
409
Collins
SJ
Acute promyelocytic leukemia: Relieving repression induces remission.
Blood
91
1998
2631
410
Chen
JD
Evans
RM
A transcriptional co-repressor that interacts with nuclear hormone receptors.
Nature
377
1995
454
411
Chen
JD
Umesono
K
Evans
RM
SMRT isoforms mediate repression and anti-repression of nuclear receptor heterodimers.
Proc Natl Acad Sci USA
93
1996
7567
412
Miller
AC
Whittaker
T
Thibault
A
Samid
D
Modulation of radiation response of human tumour cells by the differentiation inducers, phenylacetate and phenylbutyrate.
Int J Radiat Biol
72
1997
211
413
Collins
AF
Pearson
HA
Giardina
P
McDonagh
KT
Brusilow
SW
Dover
GJ
Oral sodium phenylbutyrate therapy in homozygous beta thalassemia: A clinical trial.
Blood
85
1995
43
414
Koken
M
Daniel
M-T
Gianni
M
Zelent
A
Licht
J
Degos
L
Varet
B
de Thé
H
Retinoic Acid, but not arsenic trioxide, degrades the PLZF/RARa fusion protein, without inducing terminal differentiation or apoptosis, in a RA-therapy resistant t(11;17) APL patient.
Oncogene
18
1999
1113
415
Ball
HJ
Li
J-Y
Zelent
A
Waxman
S
Licht
JD
Binding of the promyelocytic leukemia zinc finger protein (PLZF) to a genomic binding site is modulated by the conserved POZ domain and phosphorylation.
Blood
90
1997
321a
(abstr, suppl 1)
416
He
LZ
Ivins
S
Zelent
A
Pandolfi
PP
Role of RARα-PLZF in the pathogenesis of acute promyelocytic leukemia.
Blood
92
1998
480a
(abstr, suppl 1)
417
Aneskievich
BJ
Fuchs
E
The A/B domain of truncated retinoic acid receptors can block differentiation and promote features of malignancy.
J Cell Sci
108
1995
195
418
Brunel
V
Sainty
D
Carbuccia
N
Arnoulet
C
Costello
R
Mozziconacci
MJ
Simonetti
J
Coignet
L
Gabert
J
Stoppa
AM
Birg
F
Lafage-Pochitaloff
M
Unbalanced translocation t(5;17) in an typical acute promyelocytic leukemia.
Genes Chromosomes Cancer
14
1995
307
419
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
420
Chan
W
Liu
Q
Borjigin
J
Busch
H
Rennert
O
Tease
L
Chan
P
Characterization of the cDNA encoding human nucleophosmin and studies of its role in normal and abnormal growth.
Biochemistry
28
1989
1033
421
Olson
O
Orrick
L
Jones
C
Busch
H
Phosphorylation of acid soluble nulceolar proteins of novikoff hepatoma ascites cells in vivo.
J Biol Chem
249
1974
2823
422
Hernandez-Verdun
D
The nucleolar organizer regions.
Biol Cell
49
1983
191
423
Hernandez-Verdun
D
Derenzeni
M
Bouteille
M
The morphological relationship in electron microscopy between NOR-silver proteins and intranuclear chromatin.
Chromsoma
85
1982
461
424
Chan
PK
Chan
FY
Morris
SW
Xie
Z
Isolation and characterization of the human nucleophosmin/B23 (NPM) gene: Identification of the YY1 binding site at the 5′ enhancer region.
Nucleic Acids Res
25
1997
1225
425
Morris
S
Kirstein
M
Valentine
M
Dittmer
K
Shapiro
D
Saltman
D
Look
T
Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in a non-Hodgkin’s lymphoma.
Science
263
1994
1281
426
Chang
J
Olson
M
Structure of the gene for rat nucleolar protein B23.
J Biol Chem
265
1990
18227
427
Chang
J
Olson
M
A single gene codes for two forms of rat nucleolar protein B23 mRNA.
J Biol Chem
264
1989
11732
428
Chan
P
Chan
F
Morris
S
Zhong
X
Isolation and characterization of the human nucleophosmin/B23 (NPM) gene: Identification of the YY1 binding site at the 5′ enhancer region.
Nucleic Acids Res
25
1997
1225
429
Lee
JH
Welch
DR
Identification of highly expressed genes in metastasis-suppressed chromosome 6/human malignant melanoma hybrid cells using subtractive hybridization and differential display.
Int J Cancer
71
1997
1035
430
Chan
P
Aldrich
M
Cook
R
Busch
H
Amino acid sequence of B23 phosphorylation site.
J Biol Chem
261
1986
1868
431
Chan
P
Chan
W
Yung
B
Cook
R
Aldrich
M
Ku
D
Goldknopf
I
Busch
H
Amino acid sequence of a specific antigenic peptide of protein B23.
J Biol Chem
261
1986
14335
432
Chang
J
Dumbar
T
Olson
M
cDNA and deduced primary structure of rat protein B23, a nucleolar protein containing highly conserved sequences.
J Biol Chem
263
1988
12824
433
Li
Y
Busch
R
Valdez
B
Busch
H
C23 interacts with B23, a putative nucleolar-localization-signal-binding protein.
Eur J Biochem
237
1996
153
434
Valdez
B
Perlaky
L
Henning
D
Saijo
Y
Chan
P
Busch
H
Identification of the nuclear and nucleolar localization signals of the protein p120 interaction with translocation protein B23.
J Biol Chem
269
1994
23776
435
Chan
P
Cross-linkage of nucleophosmin in tumor cells by nitrogen mustard.
Cancer Res
49
1989
3271
436
Szebeni
A
Herrera
J
Olson
M
Interaction of nucleolar protein B23 with peptides related to nuclear localization signals.
Biochemistry
34
1995
8037
437
Marasco
W
Szilvay
A
Kalland
K
Helland
D
Reyes
H
Walter
R
Spatial association of HIV-1 tat protein and the nucleolar transport protein B23 in stably transfected Jurkat T-cells.
Arch Virol
139
1994
133
438
Adachi
Y
Copeland
T
Hatanake
M
Oroszlan
S
Nucleolar targeting signal of Rex protein of human T-cell leukemia virus type I specifically binds to nucleolar shuttle protein B23.
J Biol Chem
268
1993
13930
439
Miyazaki
Y
Takamatsu
T
Nosaka
T
Fujita
S
Martin
T
Hatanaka
M
The cytotoxicity of human immunodeficiency virus type 1 Rev: Implications for its interaction with the nucleolar protein B23.
Exp Cell Res
219
1995
93
440
Chang
JH
Lin
JY
Wu
MH
Yung
BY
Evidence for the ability of nucleophosmin/B23 to bind ATP.
Biochem J
329
1998
539
441
Peculis
B
Gall
J
Localization of the nucleolar protein NO38 in amphibian oocytes.
J Cell Biol
116
1992
1
442
Feuerstein
N
Mond
J
Kinchington
P
Hickey
R
Lindsberg
M
Hay
I
Ruyechan
W
Evidence for DNA binding activity of numatrin (B23) a cell cycle-regulated nuclear matrix protein.
Biochim Biophys Acta
1087
1990
127
443
Wang
D
Baumann
A
Szebeni
A
Olson
M
The nucleic acid binding activityof nucleolar protein B23.1 resides in its carboxyl-terminal.
J Biol Chem
269
1994
30994
444
Takemura
M
Ohta
N
Furuichi
Y
Takahashi
T
Yoshida
S
Olson
M
Umekawa
H
Stimulation of calf thymus DNA polymerase alpha activity by nucleolar protein B23.
Biochem Biophys Res Commun
199
1994
46
445
Chan
P
Chan
F
Nucleophosmin/B23 (NPM) oligomer is a major and stable entity in HeLa cells.
Biochim Biophys Acta
1262
1995
37
446
Liu
Q
Chan
P
Formation of nucleophosmin/B23 oligomers requires both the amino and the carboxyl-terminal domains of the protein.
Eur J Biochem
200
1991
715
447
Spector
D
Ochs
R
Busch
H
Silver staining, immunofluorescence, and immunoelectron microscope localization of nucleolar phosphoproteins B23 and C23.
Chromosoma
90
1984
139
448
Smetana
K
Ochs
R
Lischwe
M
Gyorkey
F
Freireich
E
Chudimael
V
Busch
H
Immunofluorescence studies on proteins B23 and C23 in nucleoli of human lymphocytes.
Exp Cell Res
152
1984
195
449
Biggiogera
M
Fakan
S
Kaufman
S
Black
A
Shaper
J
Busch
H
Simultaneous immunoelectron microscopic visualization of protein B23 and C23 distribution in the HeLa cell nucleolus.
J Histochem Cytochem
37
1989
1371
450
Herrera
J
Savkur
R
Olson
M
The ribonuclease activity of nucleolar protein B23.
Nucleic Acids Res
23
1995
3974
451
Herrera
J
Correia
J
Jones
A
Olson
M
Sedimentation analyses of the salt- and divalent metal ion-induced oligomerization of nucleolar protein B23.
Biochemistry
35
1996
2668
452
Borggrefe
T
Wabl
M
Akhmedov
AT
Jessberger
R
A B-cell-specific DNA recombination complex.
J Biol Chem
273
1998
17025
453
Laskey
RA
Mills
AD
Philpott
A
Leno
GH
Dilworth
SM
Dingwall
C
The role of nucleoplasmin in chromatin assembly and disassembly.
Phil Trans R Soc Lond B Biol Sci
339
1993
263
454
Chen
H
Li
B
Workman
JL
A histone-binding protein, nucleoplasmin, stimulates transcription factor binding to nucleosomes and factor-induced nucleosome disassembly.
EMBO J
13
1994
380
455
Szebeni
A
Mehrotra
B
Baumann
A
Adam
SA
Wingfield
PT
Olson
MO
Nucleolar protein B23 stimulates nuclear import of the HIV-1 Rev protein and NLS-conjugated albumin.
Biochemistry
36
1997
3941
456
Li
YP
Protein B23 is an important human factor for the nucleolar localization of the human immunodeficiency virus protein Tat.
J Virol
71
1997
4098
457
Derenzini
M
Sirri
V
Trere
D
Ochs
R
The quantity of nucleolar proteins nucleolin and protein B23 is related to cell doubling time in human cancer cells.
Lab Invest
73
1995
497
458
Derenzini
M
Sirri
V
Pession
A
Trere
D
Roussel
P
Ochs
R
Hernandez-Verdun
D
Quantitative changes of the two major AgNOR proteins, nucleolin and protein B23, related to stimulation of rDNA transcription.
Exp Cell Res
219
1995
276
459
Busch
H
Busch
R
Black
A
Chan
P
Chatterjee
A
Durban
E
Freeman
J
Ochs
R
Reichlin
M
Tan
E
Ross
B
Yaneva
M
Novel nucleolar antigens in autoimmune disease.
J Rheumatol
14
1987
70
(suppl 13)
460
Feuerstein
N
Spiegel
S
Mond
J
The nuclear matrix protein, numatrin (B23), is associated with growth factor-induced mitogenesis in Swiss 3T3 fibroblasts and with T-lymphocyte proliferation stimulated by lectins and anti-T cell antigen receptor antibody.
J Cell Biol
107
1988
1629
461
Kondo
T
Minamino
N
Nagamura-Inoue
T
Matsumoto
M
Taniguchi
T
Tanaka
N
Identification and characterization of nucleophosmin/B23/numatrin which binds the anti-oncogenic transcription factor IRF-1 and manifests oncogenic activity.
Oncogene
15
1997
1275
462
Inouye
C
Seto
E
Relief of YY1-induced transcriptional repression by protein-protein interaction with the nucleolar phosphoprotein B23.
J Biol Chem
259
1994
6506
463
Shi
Y
Lee
J
Galvin
K
Everything you have ever wanted to know about Yin Yang 1. . . . ...
Biochem Biophys Acta
1332
1997
F49
464
Hsu
CY
Yung
BY
Down-regulation of nucleophosmin/B23 during retinoic acid-induced differentiation of human promyelocytic leukemia HL-60 cells.
Oncogene
16
1998
915
465
Sirri
V
Roussel
P
Gendron
MC
Hernandez-Verdun
D
Amount of the two major Ag-NOR proteins, nucleolin, and protein B23 is cell-cycle dependent.
Cytometry
28
1997
147
466
De Angelis
PM
Stokke
T
Clausen
OP
NO38 expression and nucleolar counts are correlated with cellular DNA content but not with proliferation parameters in colorectal carcinomas.
Mol Pathol
50
1997
201
467
Feuerstein
N
Chan
P
Mond
J
Identification of numatrin, the nuclear matrix protein associated with induction of mitogenesis, as the nucleolar protein B23.
J Biol Chem
263
1988
10608
468
Zatsepina
OV
Todorov
IT
Philipova
RN
Krachmarov
CP
Trendelenburg
MF
Jordan
EG
Cell cycle-dependent translocations of a major nucleolar phosphoprotein, B23, and some characteristics of its variants.
Eur J Cell Biol
73
1997
58
469
Peter
M
Nakagawa
J
Doree
M
Labbe
J
Nigg
E
Identification of major nucleolar proteins as candidate mitotic substrates of cdc2 kinase.
Cell
6
1990
791
470
Tawfic
S
Goueli
S
Olson
M
Ahmed
K
Androgenic regulation of the expression and phosphorylation of prostatic nucleolar protein B23.
Cell Mol Biol Res
39
1993
43
471
Tawfic
S
Olson
M
Ahmed
K
Role of protein phosphorylation in post-translational regulation of protein B23 during programmed cell death in the prostate gland.
J Biol Chem
270
1995
21009
472
Ramsamooj
P
Notario
V
Dritschilo
A
Modification of nucleolar protein B23 after exposure to ionizing radiation.
Radiat Res
143
1995
158
473
Yung
B
Busch
R
Busch
H
Mauger
A
Chan
P
Effects of actinomycin D analogs on nucleolar phosphoprotein B23 (39 kDA/pI5.1).
Biochem Pharmacol
34
1985
4059
474
Chan
P
Aldrich
M
Yung
B
Nucleolar protein B23 translocation after doxorubicin treatment in murine tumor cells.
Cancer Res
47
1987
3798
475
Yung
B
Bor
A
Chan
P
Short exposure to actinomycin D induces “reversible” translocation of protein B23 as well as “reversible” inhibition of cell growth and RNA synthesis in HeLa cells.
Cancer Res
50
1990
5987
476
Yung
B
Yang
Y
Bor
A
Nucleolar protein B23 translocation after deferoxamine treatment in a human leukemia cell line.
Int J Cancer
48
1991
779
477
Lui
C
Chan
P
Fung
K
Choy
Y
Lee
C
Effects of hyperthermia on the nucleolar proteins in tumor cells.
Cancer Lett
70
1993
129
478
Chan
P
Characterization and cellular localization of nucleophosmin/B23 in HeLa cells treated with selected cytotoxic agents (studies of B23-translocation mechanism).
Exp Cell Res
203
1992
174
479
Dudnik
O
Zatsepina
O
The behavior of nucleolar proteins under the conditions of the reversible three-dimensional separation of the structural components of the nucleolus.
Tsitologia
37
1995
126
480
Hui
EKW
Yang
YH
Yung
YM
Schedule-dependent sphinganine potentiation of retinoic acid-induced differentiation, cell growth inhibition, and nucleophosmin translocation in a human leukemia cell line (HL-60).
Exp Hematol
20
1992
454
481
Yung
B
Chang
F
Luo
K
Dipyridamole enhancement of doxorubicin-induced translocation of nucleophosmin and inhibition of cell growth in HL-60 cells.
Int J Cancer
49
1991
592
482
Yung
B
Busch
H
Chan
P
Effects of luzoproteins on protein B23 translocation and ribosomal RNA synthesis in HeLa cells.
Cancer Res
46
1986
922
483
Chan
P
Qi
Y
Amley
J
Koller
C
Quantitation of the nucleophosmin/B23-translocation using imaging analysis.
Cancer Lett
100
1996
191
484
Rimokh
R
Magaud
J-P
Berger
F
Coiffier
B
Germain
D
Mason
D
A translocation involving a specific breakpoint (q35) on chromosome 5 is characteristic of anaplastic large cell lymphoma (‘Ki-1 lymphoma’).
Br J Haematol
71
1989
31
485
Ebrahim
S
Ladanyi
M
Desai
S
Offit
K
Jhanwar
S
Filippa
D
Lieberman
P
Chaganti
R
Immunohistochemical, molecular, and cytogenetic analysis of a consecutive series of 20 peripheral T-cell lymphomas and lymphomas of uncertain lineage, including 12 Ki-1 positive lymphomas.
Genes Chromosomes Cancer
2
1990
27
486
Bullrich
F
Morris
S
Hummel
M
Pileri
S
Stein
H
Croce
C
Nucleophosmin (NPM) gene rearrangements in Ki-1-positive lymphomas.
Cancer Res
54
1994
2873
487
Ladanyi
M
Cavalchire
G
Morris
S
Downing
J
Filippa
D
Reverse transcriptase polymerase chain reaction for the Ki-1 anaplastic large cell lymphoma-associated t(2;5) translocation in Hodgkin’s disease.
Am J Pathol
145
1994
1296
488
Orscheschek
K
Merz
H
Hell
J
Binder
T
Barteis
H
Feller
A
Large-cell anaplastic lymphoma-specific translocation (t[2;5] [p23;q35]) in Hodgkin’s disease: Indication of a common pathogenesis?
Lancet
345
1995
87
489
Wellman
A
Otsuki
T
Vogelbruch
M
Clark
H
Jaffe
E
Raffeld
M
Analysis of the t(2;5) (p23;q35) translocation by reverse transcription-polymerase chain reaction in CD30+ anaplastic large cell lymphomas, in other non-Hodgkin’s lymphomas of T-cell phenotype, and in Hodgkin’s disease.
Blood
86
1995
2321
490
Shiota
M
Nakamura
S
Ichinohasama
R
Abe
M
Akagi
T
Takeshita
M
Mori
N
Fujimoto
J
Miyauchi
J
Mikata
A
Nanba
K
Takami
T
Yamabe
H
Takano
Y
Izumo
T
Nagatani
T
Mohri
N
Nasu
K
Satoh
H
Katano
H
Fujimoto
J
Yamamoto
T
Mori
S
Anaplastic large cell lymphomas expressing the novel chimeric protein p80NPM/ALK: A distinct clinicopathologic entity.
Blood
86
1995
1954
491
Elmberger
G
Lozano
M
Weisenburger
D
Sanger
W
Chan
W
Transcripts of the npm-alk fusion gene in anaplastic large cell lymphoma, Hodgkin’s disease and reactive lymphoid lesions.
Blood
86
1995
3517
492
Downing
J
Shurtleff
S
Zielenska
M
Curcio-Brint
A
Behm
F
Head
D
Sandlund
J
Weisenburger
D
Kossakowska
A
Thorner
P
Lorenzana
A
Ladanyi
M
Morris
S
Molecular detection of the (2;5) translocation of non-Hodgkin’s lymphoma by reverse transcriptase-polymerase chain reaction.
Blood
85
1995
3416
493
Weiss
L
Lopategui
J
Sun
L
Kamel
O
Koo
C
Glackin
C
Absence of the t(2;5) in Hodgkin’s disease.
Blood
85
1995
2845
494
Lopategui
J
Sun
L
Chan
J
Gaffey
M
Frierson
H
Jr
Glackin
C
Weiss
L
Low frequency association of the t(2;5) (p23;q35) chromosomal translocation with CD30+ lymphomas from American and Asian patients.
Am J Pathol
146
1995
323
495
Yee
H
Ponzoni
M
Merson
A
Goldstein
M
Scarpa
A
Chilosi
M
Menestrina
F
Pittaluga
S
De Wolf-Peeters
C
Shiota
M
Mori
S
Frizzera
G
Inghirami
G
Molecular characterization of the t(2;5) (p23;q35) translocation in anaplastic large cell lymphoma (Ki-1) and Hodgkin’s disease.
Blood
87
1996
1081
496
Iwahara
T
Fujimoto
J
Wen
D
Cupples
R
Bucay
N
Arakawa
T
Mori
S
Ratzkin
B
Yamamoto
T
Molecular characterization of ALK, a receptor tyrosine kinase expressed specifically in the nervous system.
Oncogene
14
1997
439
497
Shiota
M
Fujimoto
J
Semba
T
Satoh
H
Yamamoto
T
Mori
S
Hyperphosphorylation of a novel 80 KDa protein-tyrosine kinase similar to Ltk in a human Ki-1 lymphoma cell line, AMS3.
Oncogene
9
1994
1567
498
Fujimoto
J
Shiota
M
Iwahara
T
Seki
N
Satoh
h
Mori
S
Yamamoto
T
Characterizationof the transforming activity of p80, a hyperphosphorylated protein in a Ki-1 lymphoma cell line with chromosomal translocation t(2;5).
Proc Natl Acad Sci USA
93
1996
4181
499
Bischof
D
Pulford
K
Mason
D
Morris
S
Role of nucleophosmin (NPM) portion of the non-Hodgkin’s lymphoma-associated NPM-anaplastic lymphoma kinase fusion protein in oncogenesis.
Mol Cell Biol
17
1997
2312
500
Mason
DY
Pulford
KA
Bischof
D
Kuefer
MU
Butler
LH
Lamant
L
Delsol
G
Morris
SW
Nucleolar localization of the nucleophosmin-anaplastic lymphoma kinase is not required for malignant transformation.
Cancer Res
58
1998
1057
501
Wellmann
A
Doseeva
V
Butscher
W
Raffeld
M
Fukushima
P
Stetler-Stevenson
M
Gardner
K
The activated anaplastic lymphoma kinase increases cellular proliferation and oncogene up-regulation in rat 1a fibroblasts.
FASEB J
11
1997
965
502
Kuefer
MU
Look
AT
Pulford
K
Behm
FG
Pattengale
PK
Mason
DY
Morris
SW
Retrovirus-mediated gene transfer of NPM-ALK causes lymphoid malignancy in mice.
Blood
90
1997
2901
503
Yoneda-Kato
N
Look
T
Kirstein
M
Valentine
M
Raimondi
S
Cohen
K
Carroll
A
Morris
S
The t(3;5)(q25.1;q34) of myelodysplastic syndrome and acute myeloid leukemia produces a novel fusion gene, NPM-MLF1.
Oncogene
10
1996
265
504
Corey
SJ
Locker
J
Oliveri
DR
Shekhter-Levin
S
Redner
RL
Penchansky
L
Gollin
SM
A non-classical translocation involving 17q12 (retinoic acid receptor alpha) in acute promyelocytic leukemia (APML) with atypical features.
Leukemia
8
1994
1350
505
Redner
RL
Corey
SJ
Rush
EA
Differentiation of t(5;17) variant acute promyelocytic leukemic blasts by all-trans retinoic acid.
Leukemia
11
1997
1014
506
Hummel
J
Wells
RA
Kamel-Reid
S
Dube
ID
Characterization of a variant pediatric APL; molecular evidence for dysregulation of RARA by recombination with a novel loocus on chromsome 5.
J Cell Biochem
56
1994
192
(suppl 18a)
507
Redner
R
Rush
L
Pollock
S
Forced expression of the NPM/RAR fusion gene inhibits differentiation of U937 cells.
Blood
88
1996
551a
(abstr, suppl 1)
508
Cheng
G-X
Meng
X-Q
Jin
X-L
Wang
L
Zhu
J
Xiong
SM
Zhu
X-H
Waxman
S
Licht
J
Zelent
A
Dong
S
Chen
G-Q
Chen
S-J
Establishment of transgenic leukemia mice models with acute promyelocytic leukemia-specific fusion genes PLZF-RARα and NPM-RARα.
Blood
92
1998
213a
(abstr, suppl 1)
509
Wells
RA
Hummel
JL
De Koven
A
Zipursky
A
Kirby
M
Dube
I
Kamel-Reid
S
A new variant translocation in acute promyelocytic leukaemia: Molecular characterization and clinical correlation.
Leukemia
10
1996
735
510
Kallajoki
M
Weber
K
Osborn
M
Ability to organize microtubules in taxol-treated mitotic Ptk2 cells goes with the SPN antigen and not with the centromere.
J Cell Sci
102
1992
91
511
Kallajoki
M
Weber
K
Osborn
M
A 210 kD nuclear matrix protein is a functional part of the mitotic spindle: A microinjection study using SPN monoclonal antibodies.
EMBO J
10
1991
3351
512
Compton
DA
Cleveland
DW
NuMA, a nuclear protein involved in mitosis and nuclear reformation.
Curr Opin Cell Biol
6
1994
343
513
He
D
Zeng
C
Brinkley
BR
Nuclear matrix proteins as structural and functional components of the mitotic apparatus.
Int Rev Cytol
162B
1995
1
514
Saredi
A
Howard
L
Compton
DA
NuMA assembles into an extensive filamentous structure when expressed in the cell cytoplasm.
J Cell Sci
109
1996
619
515
Compton
DA
Szilak
I
Cleveland
DW
Primary structure of NuMA, an intranuclear protein that defines a novel pathway for segregation of proteins at mitosis.
J Cell Biol
116
1992
1395
516
Lyderson
B
Kao
F
Pettijohn
D
Expression of genes coding for non-histone chromosomal proteins in human/chinese hamster cell hybrids: An electrophoretic analysis.
J Biol Chem
255
1980
3002
517
Lyderson
B
Pettijohn
D
Human-specific nuclear protein that associates with the polar region of the mitotic apparatus: Distribution in a human/hamster hybrid cell.
Cell
22
1980
489
518
Sparks
CA
Bangs
PL
McNeil
GP
Lawrence
JB
Fey
EG
Assignment of the nuclear mitotic apparatus protein NuMA gene to human chromosome 11q13.
Genomics
17
1993
222
519
Tang
TK
Tang
CJ
Chen
YL
Wu
CW
Nuclear proteins of the bovine esophageal epithelium. II. The NuMA gene gives rise to multiple mRNAs and gene products reactive with monoclonal antibody W1.
J Cell Sci
104
1993
249
520
Tang
TK
Tang
CJ
Chao
YJ
Wu
CW
Nuclear mitotic apparatus protein (NuMA): Spindle association, nuclear targeting and differential subcellular localization of various NuMA isoforms.
J Cell Sci
107
1994
1389
521
Yang
CH
Lambie
EJ
Snyder
M
NuMA: An unusually long coiled-coil related protein in the mammalian nucleus.
J Cell Biol
116
1992
1303
522
Maekawa
T
Kuriyama
R
Primary structure and microtubule-interacting domain of the SP-H antigen: A mitotic MAP located at the spindle pole and characterized as a homologous protein to NuMA.
J Cell Sci
105
1993
589
523
Parry
DA
NuMA/centrophilin: Sequence analysis of the coiled-coil rod domain.
Biophys J
67
1994
1203
524
Harborth
J
Weber
K
Osborn
M
Epitope mapping and direct visualization of the parallel, in-register arrangement of the double-stranded coiled-coil in the NuMA protein.
EMBO J
14
1995
2447
525
Compton
DA
Cleveland
DW
NuMA is required for the proper completion of mitosis.
J Cell Biol
120
1993
947
526
Hsu
HL
Yeh
NH
Dynamic changes of NuMA during the cell cycle and possible appearance of a truncated form of NuMA during apoptosis.
J Cell Sci
109
1996
277
527
Yang
CH
Snyder
M
The nuclear-mitotic apparatus protein is important in the establishment and maintenance of the bipolar mitotic spindle apparatus.
Mol Biol Cell
3
1992
1259
528
Gueth-Hallonet
C
Weber
K
Osborn
M
NuMA: A bipartite nuclear location signal and other functional properties of the tail domain.
Exp Cell Res
225
1996
207
529
Saredi
A
Howard
L
Compton
DA
Phosphorylation regulates the assembly of NuMA in a mammalian mitotic extract.
J Cell Sci
110
1997
1287
530
Maekawa
T
Leslie
R
Kuriyama
R
Identification of a minus end-specific microtubule-associated protein located at the mitotic poles in cultured mammalian cells.
Eur J Cell Biol
54
1991
255
531
Tousson
A
Zeng
C
Brinkley
B
Valdivia
M
Centrophilin: A novel mitotic spindle protein involved in nicrotubule nucleation.
J Cell Biol
112
1991
427
532
Kallajoki
M
Harborth
J
Weber
K
Osborn
M
Microinjection of a monoclonal antibody against SPN antigen, now identified by peptide sequences as the NuMA protein, induces micronuclei in PtK2 cells.
J Cell Sci
104
1993
139
533
Gaglio
T
Saredi
A
Compton
DA
NuMA is required for the organization of microtubules into aster-like mitotic arrays.
J Cell Biol
131
1995
693
534
Merdes
A
Ramyar
K
Vechio
JD
Cleveland
DW
A complex of NuMA and cytoplasmic dynein is essential for mitotic spindle assembly.
Cell
87
1996
447
535
Gaglio
T
Saredi
A
Bingham
JB
Hasbani
MJ
Gill
SR
Schroer
TA
Compton
DA
Opposing motor activities are required for the organization of the mammalian mitotic spindle pole.
J Cell Biol
135
1996
399
536
Price
C
Pettijohn
D
Distribution of the nuclear mitotic apparatus protein (NuMa) during mitosis and nuclear assembly.
Exp Cell Res
166
1986
295
537
Kempf
T
Bischoff
FR
Kalies
I
Ponstingl
H
Isolation of human NuMA protein.
FEBS Lett
354
1994
307
538
Zeng
C
He
D
Brinkley
BR
Localization of NuMA protein isoforms in the nuclear matrix of mammalian cells.
Cell Motil Cytoskeleton
29
1994
167
539
Zeng
C
He
D
Berget
SM
Brinkley
BR
Nuclear-mitotic apparatus protein: A structural protein interface between the nucleoskeleton and RNA splicing.
Proc Natl Acad Sci USA
91
1994
1505
540
Tsutsui
K
Tsutsui
K
Okada
S
Waterai
S
Seki
S
Yasuda
T
Shomori
T
Identification and characterization of a nuclear scaffold protein that binds the matrix attachment region DNA.
J Biol Chem
268
1993
12886
541
Luderus
ME
den Blaauwen
JL
de Smit
OJ
Compton
DA
van Driel
R
Binding of matrix attachment regions to lamin polymers involves single-stranded regions and the minor groove.
Mol Cell Biol
14
1994
6297
542
Zweyer
M
Riederer
BM
Ochs
RL
Fackelmayer
FO
Kohwi-Shigematsu
T
Bareggi
R
Narducci
P
Martelli
AM
Association of nuclear matrix proteins with granular and threaded nuclear bodies in cell lines undergoing apoptosis.
Exp Cell Res
230
1997
325
543
Casiano
C
Martin
S
Green
D
Tan
E
Selective cleavage of nuclear autoantigens during CD95 (Fas/APO-1)-mediated T cell apoptosis.
Exp Med
184
1996
765
544
Sodja
C
Walker
PR
Brown
DL
Chaly
N
Unique behaviour of NuMA during heat-induced apoptosis of lymphocytes.
Biochem Cell Biol
75
1997
399
545
Hirata
H
Takahashi
A
Kobayashi
S
Yonehara
S
Sawai
H
Okazaki
T
Yamamoto
K
Sasada
M
Caspases are activated in a branched protease cascade and control distinct downstream processes in Fas-induced apoptosis.
J Exp Med
187
1998
587
546
Gueth-Hallonet
C
Weber
K
Osborn
M
Cleavage of the nuclear matrix protein NuMA during apoptosis.
Exp Cell Res
233
1997
21
547
Sanghavi
DM
Thelen
M
Thornberry
NA
Casciola-Rosen
L
Rosen
A
Caspase-mediated proteolysis during apoptosis: Insights from apoptotic neutrophils.
FEBS Lett
422
1998
179
548
Wells
RA
Catzavelos
C
Kamel-Reid
S
Fusion of retinoic acid receptor alpha to NuMA, the nuclear mitotic apparatus protein, by a variant translocation in acute promyelocytic leukaemia.
Nat Genet
17
1997
109
549
Warrell
RPJ
He
LZ
Richon
V
Calleja
E
Pandolfi
PP
Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase.
J Natl Cancer Inst
90
1998
1621
550
Burger
C
Wick
M
Muller
R
Lineage-specific regulation of cell cycle gene expression in differentiating myeloid cells.
J Cell Sci
107
1994
2047
551
Pahl
HL
Rosmarin
AG
Tenen
DG
Characterization of the myeloid-specific CD11b promoter.
Blood
79
1992
865
552
Rosmarin
AG
Levy
R
Tenen
DG
Cloning and analysis of the CD18 promoter.
Blood
79
1992
2598
553
Hickstein
DD
Baker
DM
Gollahon
KA
Back
AL
Identification of the promoter of the myelomonocytic leukocyte integrin CD11b.
Proc Natl Acad Sci USA
89
1992
2105
554
Di Noto
R
Schiavone
EM
Ferrara
F
Manzo
C
Lo Pardo
C
Del Vecchio
L
Expression and ATRA-driven modulation of adhesion molecules in acute promyelocytic leukemia.
Leukemia
8
1994
S71
(suppl 2)
555
Grande
A
Manfredini
R
Tagliafico
EAUBR
Pizzanelli
M
Papa
S
Zucchini
P
Bonsi
L
Bagnara
G
Torelli
U
Ferrari
S
All-trans-retinoic acid induces simultaneously granulocytic differentiation and expression of inflammatory cytokines in HL-60 cells.
Exp Hematol
23
1995
117
556
Herwig
S
Su
Q
Zhang
W
Ma
Y
Tempst
P
Distinct temporal patterns of defensin mRNA regulation during drug-induced differentiation of human myeloid leukemia cells.
Blood
87
1996
350
557
Gianni
M
Terao
M
Zanotta
S
Barbui
T
Rambaldi
A
Garattini
E
Retinoic acid and granulocyte colony-stimulating factor synergistically induce leukocyte alkaline phosphatase in acute promyelocytic leukemia cells.
Blood
83
1994
1909
558
Garattini
E
Gianni
M
Leukocyte alkaline phosphatase a specific marker for the post-mitotic neutrophilic granulocyte: Regulation in acute promyelocytic leukemia.
Leuk Lymphoma
23
1996
493
559
Lee
MO
Liu
Y
Zhang
XK
A retinoic acid response element that overlaps an estrogen response element mediates multihormonal sensitivity in transcriptional activation of the lactoferrin gene.
Mol Cell Biol
15
1995
4194
560
Matikainen
S
Tapiovaara
H
Vaheri
A
Hurme
M
Activation of interleukin-1 beta gene expression during retinoic acid-induced granulocytic differentiation of promyeloid leukemia cells.
Cell Growth Differ
5
1994
975
561
Dubois
C
Schlageter
MH
de Gentile
A
Balitrand
N
Toubert
ME
Krawice
I
Fenaux
P
Castaigne
S
Najean
Y
Degos
L
Chomienne
C
Modulation of IL-8, IL-1 beta, and G-CSF secretion by all-trans retinoic acid in acute promyelocytic leukemia.
Leukemia
8
1994
1750
562
Hsu
HC
Yang
K
Kharbanda
S
Clinton
S
Datta
R
Stone
RM
All-trans retinoic acid induces monocyte growth factor receptor (c-fms) gene expression in HL-60 leukemia cells.
Leukemia
7
1993
458
563
Tkatch
LS
Rubin
KA
Ziegler
SF
Tweardy
DJ
Modulation of human G-CSF receptor mRNA and protein in normal and leukemic myeloid cells by G-CSF and retinoic acid.
J Leukoc Biol
57
1995
964
564
Dipietrantonio
A
Hsieh
TC
Wu
JM
Differential effects of retinoic acid (RA) and N-(4-hydroxyphenyl) retinamide (4-HPR) on cell growth, induction of differentiation, and changes in p34cdc2, Bcl-2, and actin expression in the human promyelocytic HL-60 leukemic cells.
Biochem Biophys Res Commun
224
1996
837
565
Davies
P
Murtaugh
M
Moore
W
Johnson
G
Lucas
D
Retinoic acid-induced expression of transglutaminase in human promyelocytic leukemia (HL-60).
J Biol Chem
260
1985
5166
566
Falanga
A
Iacoviello
L
Evangelista
V
Belotti
D
Consonni
R
D’Orazio
A
Robba
L
Donati
MB
Barbui
T
Loss of blast cell procoagulant activity and improvement of hemostatic variables in patients with acute promyelocytic leukemia administered all-trans-retinoic acid.
Blood
86
1995
1072
567
Koyama
T
Hirosawa
S
Kawamata
N
Tohda
S
Aoki
N
All-trans retinoic acid upregulates thrombomodulin and downregulates tissue-factor expression in acute promyelocytic leukemia cells: Distinct expression of thrombomodulin and tissue factor in human leukemic cells.
Blood
84
1994
3001
568
Tapiovaara
H
Matikainen
S
Hurme
M
Vaheri
A
Induction of differentiation of promyelocytic NB4 cells by retinoic acid is associated with rapid increase in urokinase activity subsequently downregulated by production of inhibitors.
Blood
83
1994
1883
569
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
570
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
571
Sternsdorf
T
Jensen
K
Zuchner
D
Will
H
Cellular localization, expression, and structure of the nuclear dot protein 52.
J Cell Biol
138
1997
435
572
Desbois
C
Rousset
R
Bantignies
F
Jalinot
P
Exclusion of Int-6 from PML nuclear bodies by binding to the HTLV-I Tax oncoprotein.
Science
273
1996
951
573
Asano
K
Merrick
WC
Hershey
JW
The translation initiation factor eIF3-p48 subunit is encoded by int-6, a site of frequent integration by the mouse mammary tumor virus genome.
J Biol Chem
272
1997
23477
574
Everett
RD
Meredith
M
Orr
A
Cross
A
Kathoria
M
Parkinson
J
A novel ubiquitin-specific protease is dynamically associated with the PML nuclear domain and binds to a herpesvirus regulatory protein.
EMBO J
16
1997
566
575
Cao
T
Duprez
E
Borden
KL
Freemont
PS
Etkin
LD
Ret finger protein is a normal component of PML nuclear bodies and interacts directly with PML.
J Cell Sci
111
1998
1319
576
Kawaguchi
Y
Van Sant
C
Roizman
B
Herpes simplex virus 1 alpha regulatory protein ICP0 interacts with and stabilizes the cell cycle regulator cyclin D3.
J Virol
71
1997
7328
577
Carlile
GW
Tatton
WG
Borden
KLB
Demonstration of a RNA-dependent nuclear interaction between the promyelocytic leukaemia protein and glyceraldehyde-3-phosphate dehydrogenase.
Biochem J
335
1998
691
578
Carvalho
T
Seeler
JS
Ohman
K
Jordan
P
Pettersson
U
Akusjarvi
G
Carmo-Fonseca
M
Dejean
A
Targeting of adenovirus E1A and E4-ORF3 proteins to nuclear matrix-associated PML bodies.
J Cell Biol
131
1995
45
579
Li
X
McNeilage
L
Whittingham
S
The nucleotide sequence of a human cDNA encoding the highly conserved nucleolar phosphoprotein B23.
Biochem Biophys Res Commun
163
1989
72
580
Zang
X
Thomis
D
Samuel
C
Isolation and characterization of a molecular cDNA clone of a human mRNA from interferon-treated cells encoding nucleolar proteins B23, numatrin.
Biochem Biophys Res Commun
164
1989
176
581
Fankhauser
C
Izaurralde
E
Adachi
Y
Wingfield
P
Laemmli
U
Specific complex of human immunodeficiency virus type I Rev and nucleolar B23 proteins: Dissociation by the Rev response element.
Mol Cell Biol
11
1991
2567
582
Dundr
M
Leno
G
Hammarskjold
M
Rekosh
D
Helga-Maria
C
Olson
M
The roles of nucleolar structure and function in the subcellular location of the HIV-1 Rev protein.
J Cell Sci
108
1995
2811
583
Feuerstein
N
Randazzo
P
In vivo and In vitro phosphorylation studies of numatrin, a cell cycle regulated nuclear protein, in insulin-stimulated NIH 3T3 HIR cells.
Exp Cell Res
194
1991
289
584
Feuerstein
N
Phosphorylation of numatrin and other nuclear proteins by cdc2 containing CTD kinase cdc3/p58.
J Biol Chem
266
1991
16200
585
Beckmann
R
Buchner
K
Jungblut
P
Eckerson
C
Weise
C
Hilbert
R
Hucho
F
Nuclear substrates of protein kinase C.
Eur J Biochem
210
1992
45
586
Chan
P
Liu
Q
Durban
E
The major phosphorylation site of nucleophosmin (B23) is phosphorylated by a nuclear kinase II.
Biochem J
270
1990
549
587
Lu
Y
Lam
C
Yung
B
Decreased accumulation and dephosphorylation of the mitosis-specific form of nucleophosmin/B23 in staurosporine-induced chromosome decondensation.
Biochem J
317
1996
321
588
Van Ness
J
Pettijohn
D
Specific attachment of NuMA protein to metaphase chromosomes: Possible function in nuclear reassembly.
J Mol Biol
171
1983
175
589
Merdes
A
Cleveland
DW
The role of NuMA in the interphase nucleus.
J Cell Sci
111
1998
71
590
Feuerstein
N
Mond
J
“Numatrin”, a nucleolar matrix protein associated with induction of proliferation in B lymphocytes.
J Biol Chem
262
1987
11389
591
Sparks
CA
Fey
EG
Vidair
CA
Doxsey
SJ
Phosphorylation of NUMA occurs during nuclear breakdown and not mitotic spindle assembly.
J Cell Sci
108
1995
3389
592
Compton
DA
Luo
C
Mutation of the predicted p34cdc2 phosphorylation sites in NuMA impair the assembly of the mitotic spindle and block mitosis.
J Cell Sci
108
1995
621
593
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
594
Huynh
KD
Bardwell
VJ
The BCL-6 POZ domain and other POZ domains interact with the co-repressors N-CoR and SMRT.
Oncogene
17
1998
2473
595
Zhu
J
Guidez
F
Owen
GI
Chew
YP
Waxman
S
Mittnacht
S
Zelent
A
Acute promyelocytic leukaemia associated PLZF protein interacts with the retinoblastoma tumour suppressor gene product.
Blood
92
1998
213a
(abstr, suppl 1)
596
Melnick
A
Westendorf
JJ
Arai
S
Lutterbach
B
Ball
HJ
Polinger
A
Licht
JD
Hiebert
SW
The PLZF leukemia protein physically and functionally interacts with the ETO leukemia protein suggesting a common link of transcriptional repression in aml.
Blood
92
1998
479a
(abstr, suppl 1)
597
De
BF
Falk
LA
Ellingsworth
LR
Ruscetti
FW
Faltynek
CR
Synergy between transforming growth factor-beta and tumor necrosis factor-alpha in the induction of monocytic differentiation of human leukemic cell lines.
Blood
75
1990
626
598
Spohn
W
Ahn
Y
Busch
R
Busch
H
Electrophoretic analysis of HeLa cell and human liver nucleolar proteins and antigens.
Cancer Invest
3
1985
307
599
Nozawa
Y
Van Belzen
N
Van der Made
A
Dinjens
W
Bosman
F
Expression of nucleophosmin/B23 in normal and neoplastic colorectal mucosa.
J Pathol
178
1996
48
600
Wesierska-Gadek
J
Penner
E
Hitchman
E
Kier
P
Sauermann
G
Nucleolar proteins B23 and C23 as target antigens in chronic graft-versus-host-disease.
Blood
79
1992
1081
601
Imai
H
Ochs
R
Kiyosawa
K
Furuta
N
Nakamura
RM
Tan
E
Nucleolar antigens and autoantibodies in hepatocellular carcinoma and other malignancies.
Am J Pathol
140
1992
859
602
Imai
H
Kiyosawa
K
Chan
E
Tan
E
Autoantibodies in viral hepatitis-related hepatocellular carcinoma.
Intervirology
35
1993
73
603
Kitamura
K
Kiyoi
H
Yoshida
H
Saito
H
Ohno
R
Naoe
T
Mutant AF-2 domain of PML-RARalpha in retinoic acid-resistant NB4 cells: Differentiation induced by RA is triggered directly through PML-RARalpha and its down-regulation in acute promyelocytic leukemia.
Leukemia
11
1997
1950
604
Grimwade
D
Gorman
P
Duprez
E
Howe
K
Langabeer
S
Oliver
F
Walker
H
Culligan
D
Waters
J
Pomfret
M
Goldstone
A
Burnett
A
Freemont
P
Sheer
D
Solomon
E
Characterization of cryptic rearrangements and variant translocations in acute promyelocytic leukemia.
Blood
90
1997
4876
605
Melnick
A
Arai
S
Polinger
A
Borden
K
Ahmad
KF
Prive
G
Licht
JD
Mutational and crystallographic analysis of the PLZF BTB/POZ domain reveals structures required for dimerization and transcriptional repression.
Blood
92
1998
309a
(abstr, suppl 1)

Author notes

Address reprint requests to Jonathan D. Licht, MD, Box 1130, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029; e-mail: jlicht@smtplink.mssm.edu.

Sign in via your Institution