pmc.ncbi.nlm.nih.gov

Noxa: at the tip of the balance between life and death

  • ️Fri Jun 02 2006

. Author manuscript; available in PMC: 2012 Feb 5.

Published in final edited form as: Oncogene. 2008 Dec;27(Suppl 1):S84–S92. doi: 10.1038/onc.2009.46

Abstract

Among all Bcl2 homology domain 3 (BH3)-only proteins known to date, APR/PMAIP1/Noxa, albeit showing weak proapoptotic potential on its own, appears to be crucial in fine-tuning cell death decisions by targeting the prosurvival molecule Mcl1 for proteasomal degradation. This event appears critical for cell death induction along the mitochondrial Bcl2-regulated apoptosis pathway in response to factor deprivation or DNA damage, presumably by sensitizing the cell toward the action of additional BH3-only protein family members. This review aims to summarize the function of Noxa in normal physiology, stress-induced cell death and tumorigenesis.

Keywords: apoptosis, BH3-only proteins, p53

Discovery, genes and transcripts

In 1990 Hijikata et al. isolated the first Noxa cDNA clone from an adult T-cell leukemia (ATL) library in search for products involved in leukemogenesis. The transcript was rapidly induced by phorbol 12-myristate 13-acetate (PMA) treatment in human peripheral blood mononuclear cells, Jurkat T acute lymphoblastic leukemia (ALL) and human embryonic lung cells (Hijikata et al., 1990) and was therefore named ATL-derived PMA-responsive gene (APR). Later on it was given the HUGO designation PMA-induced protein 1 (PMAIP1). However, the function of this protein remained unknown for another decade.

APR/PMAIP1 was then rediscovered in a differential display approach using mRNA from γ-irradiated wild-type (wt) and IRF-1/p53 double-deficient mouse embryonic fibroblasts (MEF). The isolated cDNA encoded a 103 amino-acid (aa) protein, which the authors termed Noxa (Latin for damage). Sequence analysis revealed that the Noxa protein contained no known structural motif, with the exception of a Bcl2 homology (BH) domain 3, putting it into the back and then steadily growing BH3-only subgroup of proapoptotic Bcl2 family members (Oda et al., 2000). The search for the human counterpart revealed APR/PMAIP1 as the putative homologue. Curiously, mouse and human Noxa differ significantly in size and the number of BH3 domains. Mouse Noxa, as well as the rat homologue, contains two BH3 domains and both are about twice as long as the human isoform (Figure 1a). Because no other mammalian sequence available in the database to date, nor chicken or zebrafish variants of the gene, nor its putative common ancestor found in Caenorhabditis elegans, CED-13, share this feature (Figure 1b), it has been suggested that this peculiarity is due a failed gene duplication/fusion event in primordial ancestor of rats and mice (Coultas et al., 2002). Noteworthy, like in mice and rats, the human gene contains three exons (Figure 1c) but the transcript encoding the human protein (NM_21127) contains only sequences from exons 1 and 3, neglecting exon 2. This exon shows no similarity with mouse Noxa exon 2 and is only found in two splice variants, termed Noxa splicing variant 1 and 2, which encode predicted proteins of 136 and 70 aa in length, respectively (Wang and Sun, 2008). Both splice variants only share the first 19 aa with Noxa (encoded by exon 1) but otherwise differ vastly from the remaining Noxa protein sequence and lack a BH3 domain. They are rapidly degraded through proteasome-dependent and -independent pathways (Wang and Sun, 2008) and their function, if any, is unknown. Thus, exon 2 of human Noxa is probably a variant exon and functionally the human Noxa gene might therefore be considered a ‘two-exon gene’ like that of cow, swine, chicken or zebrafish.

Figure 1.

Figure 1

(a) Sequence comparison of the human, mouse and rat Noxa protein. The mitochondrial targeting sequence (MTD) and the Bcl2 homology domain 3 (BH3) domains (A- and B motif) are shown in bold, respectively. (b) Sequence alignment of BH3 domains from ancestral Egl-1, related CED-13 and Noxa from different species. (c) Noxa gene structure, transcription factor binding sites and reported mRNA transcripts. Untranslated regions and intronic sequences are shown in white, coding sequence in black.

Regulation of Noxa expression

Initial observations indicated that Noxa is a primary p53-response gene. Noxa mRNA is rapidly induced after adenovirus-mediated introduction of p53 into MEF derived from p53−/− or wt mice and in wt thymocytes subjected to γ-irradiation but in not p53−/− controls. Promoter analysis of human Noxa revealed the presence of a bona fide p53 response element 195 bp upstream of the transcriptional start site (Figure 1c). Northern blot analysis performed on mRNA isolated from various tissues of adult mice revealed constitutive low-level Noxa expression in brain, thymus, spleen, lung, kidney and testis as well as the intestinal tract (Oda et al., 2000). Whole-body γ-irradiation induced Noxa mRNA in the thymus, spleen, jejunum and transverse colon (Oda et al., 2000; Fei et al., 2002). Interestingly, in the latter two organs, induction of Noxa occurred efficiently also in the absence of p53. This indicated that, in certain tissues, Noxa induction in response to DNA damage can occur in a p53-independent manner. Along the same line, Noxa induction was also observed in response to HDM2 inhibition in p53−/− HCT116 colon cancer cells (Lau et al., 2008) and in response to oncogenic stress, caused by E1a overexpression in p53-deficient Saos-2 cells (Flinterman et al., 2005). Although speculative, this suggests a possible tumor suppressor function for Noxa independent of the p53 status of a cell, maybe as a downstream target of p73. Consistently, the transactivating isoform of p73 was shown to be able to induce transcription of this BH3-only protein and cell death (Flinterman et al., 2005).

Not surprisingly, p53 and its relatives are not the only regulators of Noxa expression, and numerous other regulators and control mechanisms have been described (Figure 2). Thus, like Puma, the second p53 target BH3-only protein gene induced after DNA damage, Noxa was also found induced under hypoxic conditions. However, although hypoxia-induced expression of Puma is mainly dependent on p53, Noxa was reported as a direct target of Hif1a, able to mediate hypoxic cell death in a p53-independent manner. A hypoxia-responsive element was identified in the Noxa promoter (Figure 1c) and a general upregulation of Noxa during hypoxia was observed in various normal and malignant tissues (Kim et al., 2004). Potentially relevant for novel treatment regimens of ischemia, elevated Noxa levels were also detected after transient ischemia experiments in vivo and liposomal delivery of Noxa antisense oligonucleotides significantly reduced infarct volumes of rat brains (Kim et al., 2004).

Figure 2.

Figure 2

Multiple signals can induce Noxa expression. Noxa transcription and protein expression can be activated by diverse apoptotic or mitogenic signals in a p53-dependent as well as -independent manner. Noxa exerts its proapoptotic function mainly by neutralizing the prosurvival Bcl2 proteins Mcl1/A1, facilitating activation of Bax and/or Bak proteins.

Another sequence-specific transcription factor implicated in regulating Noxa expression is E2f1. Binding of viral oncoproteins such as human papillomavirus protein E7 or adenoviral E1a to the retinoblastoma gene product Rb causes deregulation of its activity entailing defective cell-cycle progression and apoptosis. Ectopic expression of E2f1 can trigger cell death not only in a p53-dependent manner (through Arf) but also in a p53-independent manner. The latter may occur through p73 and/or by direct upregulation of several BH3-only proteins, including Noxa (Hershko and Ginsberg, 2004). In fact, the Noxa gene promoter contains a bona fide E2f1-binding site (Figure 1c) that allows transcription of this BH3-only protein in a p53-independent manner.

Remembering that the human Noxa homologue, APR/PMAIP1, was initially discovered as a PMA-responsive gene in malignant T cells, Alves et al. (2006) studied regulation and function of Noxa in response to PMA and mitogenic stimulation with anti-CD3 and anti-CD28 crosslinking antibodies in mature human T cells. These treatments prompted proliferation and, somewhat counterintuitive, a strong upregulation of Noxa mRNA and protein. The response was independent of p53, but interfering with protein kinase C (PKC) signaling using pharmacological inhibitors abrogated Noxa induction, implicating novel PKC family members, presumably T-cell-selective PKC, in signal transduction downstream of the T-cell receptor in the regulation of Noxa expression (Alves et al., 2006). In this context it is worth mentioning that Noxa is induced by the common γ-chain of the receptors for cytokines interleukin (IL)-7 and IL-15, although the underlying molecular mechanisms are not well understood (Alves et al., 2006).

Although studying the molecular mechanism how immunological memory T cells might escape cell death after antigen clearance, Yamashita et al. (2008) observed that the polycomb group (PcG) gene Bmi1 represses Noxa gene expression. PcG genes control transcription by remodeling chromatin structures by constituting polycomb repressive complexes enabling DNA methylation at CpG islands (Wang et al., 2004). Loss of Bmi1 did not interfere with memory T-cell proliferation but excess cell death was observed, which correlated with increased expression of several proapoptotic genes including Bax, Noxa and Puma, in line with the capacity of Bmi1 to repress the Ink4a/Arf gene locus and subsequent p53 signaling. However, neither loss of the Ink4a/Arf locus nor loss of p53 rescued T helper 2 (Th2) memory cell generation in the absence of one or both alleles of Bmi1, and Noxa expression was still elevated in these compound mutant memory T cells. Subsequent analysis demonstrated that Bmi1 was required for DNA CpG methylation of the Noxa gene locus (Yamashita et al., 2008). Indeed, compared to wt Th2 cells, trimethylation of histone H3-K27 and methylation of CpG islands of the Noxa gene promoter were reduced in Bmi1-deficient Th2 memory cells. Interestingly, binding of the Bmi1/PcG complex to the Noxa gene locus itself depended on the methylation status of the promoter because knockdown of the methyltransferase Dnmt1 caused decreased binding of Bmi1 to the Noxa promoter and increased mRNA expression (Yamashita et al., 2008).

Another interesting observation concerning Noxa regulation was recently made in human ALL. We discovered that Noxa mRNA expression, as detected by whole-genome expression profiling, was repressed in children with ALL undergoing systemic glucocorticoid (GC) monotherapy (Schmidt et al., 2006; Ploner et al., 2008). Subsequent analyses revealed that GC represses not only Noxa mRNA but also its protein in a proteasome-dependent manner. This phenomenon may have clinical relevance because functional analyses suggested that Noxa repression by GC paradoxically interferes with extent and kinetics of GC-induced apoptosis (Ploner et al., submitted for publication).

Molecular mechanisms of Noxa-induced cell death

Apoptosis induction and subcellular localization

Overexpression of mouse Noxa in HeLa cervical carcinoma and other cancer cells causes significant apoptosis, which is associated with its preferential localization to mitochondria (Oda et al., 2000). Apoptosis induced by Noxa requires a functional BH3 domain, and is accompanied by cytochrome c release and subsequent caspase activation. No binding of Noxa to proapoptotic Bax or Bak was reported so far, indicating that Noxa promotes Bax-mediated mitochondrial dysfunction indirectly by inhibiting antiapoptotic members of the Bcl2 family (Oda et al., 2000; Shibue et al., 2003).

Overexpressed Noxa protein localizes preferentially to mitochondria, as assessed by immunostaining (Oda et al., 2000). Interestingly, Noxa seems to require an intact BH3 domain not only for cell killing but also for its mitochondrial localization, suggesting that its association with this organelle may be secondary to its interaction with the prosurvival Bcl2-like molecules that it antagonizes. However, expression of a human GFP/Noxa fusion protein and truncation mutants thereof in HeLa cells revealed that aa 41–50 may constitute an bona fide mitochondrial targeting domain (MTD; Figure 1a; Seo et al., 2003). This sequence is conserved between human, mouse, rat and chicken Noxa, and a human Noxa construct lacking the MTD domain but still containing the BH3 domain was unable to induce cell death presumably due to mislocalization (Seo et al., 2003). However, because mitochondrial localization of mouse Noxa was also prevented by replacement mutations in the two BH3 domains (without altering MTD), both domains might be required for proper mitochondrial targeting. Alternatively, given the small size of human Noxa and the fact that this deletion starts immediately C-terminal to the BH3 domain, it may impair overall conformation and its ability to interact with Bcl2 and its homologues. This, rather than the destruction of a putative mitochondrial localization signal, might then cause inappropriate subcellular localization. In any rate, because cell biological data on endogenous Noxa protein expression are still lacking, its subcellular localization, before and after cell death induction, remains to be determined.

Molecular basis for the limited proapoptotic potential of Noxa

Although Noxa significantly contributes to apoptosis induced by p53 overexpression or DNA damage, in diverse cells such as Saos-2, BAF-3 pre-B cells or MEF, its apoptotic potential, when overexpressed on its own, appears to be quite variable (Oda et al., 2000; Schuler et al., 2003). Thus, Noxa overexpression per se entailed significant cell death in some systems (Oda et al., 2000), but in many other cell types, including MEF or human CEM T ALL cells, it proved poorly apoptotic (Chen et al., 2005; Ploner et al., submitted for publication).

Several possibilities may explain these observations (see also review by DCS Huang and P Bouillet, in this issue on page S128). Compared to other BH3-only proteins Noxa shows the most restricted potential to neutralize prosurvival Bcl2 molecules. Although Bim and Puma can bind all survival Bcl2 proteins with comparable affinity, Noxa selectively binds to Mcl1 and, with lower affinity, Bfl1/A1 (Chen et al., 2005). The molecular basis for this selectivity depends on critical aa residues in the amphipathic α-helical BH3 domain contacting key residues in the hydrophobic groove of the antiapoptotic Bcl2-like prosurvival proteins, formed by parts of the BH1, BH2 and BH3 domains. As shown by high-resolution structure analyses of Bclxl/Bim complexes, binding of Bim to Bclxl depends on the interaction of hydrophobic pockets, located in the binding groove of Bclxl, with four conserved hydrophobic residues in the BH3 domain of Bim (Liu et al., 2003). Replacement of one or more of these critical residues interferes with the binding specificity and affinity of the whole BH3-only protein. This suggests that the binding properties of a given BH3 molecule depend only on the nature of its BH3 domain and hence exchange of the critical residues leads to altered binding specificity of the whole molecule. In fact, exchange of only two aa in the BH3 domain of human Noxa, that is, K35E and F32I (also called Noxa m3 mutant), increased its binding efficiency for Bclxl more than 100-fold (Chen et al., 2005). Moreover, exchange of the key contact residues of one BH3-only molecule into another transmits the binding characteristic of the donor BH3 molecule. When the Noxa BH3 domain was grafted into Bim (that usually shows high proapoptotic potential due to its capacity to bind all Bcl2 prosurvival molecules), the hybrid lost the cell death inducing potential of the BimS isoform that was used in this experiment. This correlated with the imposed binding capacity restricted to Mcl1. The same was true for BimS chimeras containing the BH3 domain of Bad, which bound Bclxl but no longer Mcl1 (Chen et al., 2005). Both mutants of BimS, however, can complement each other in promoting apoptosis. These experiments support the hypothesis that the nature of BH3 domains defines binding specificity and affinity of the whole BH3-only molecule and that mitochondrial cell death ensues when all types of Bcl2-like prosurvival molecules present in a given cell type are neutralized by BH3-only proteins. Alternative modes of BH3-only protein-mediated apoptosis are discussed in more detail by Letai and co-workers in this issue of Oncogene.

Investigations into the contribution of Noxa and Puma to p53-dependent apoptosis uncovered that NIH3T3 or MEF, transformed with adenovirus E1a, died in the presence of transgenic Noxa or Puma, whereas wt cells were protected against Noxa- but not Puma-induced cell death (Shibue et al., 2006). In E1a-transformed cells Noxa enhanced activation and oligomerization of Bax but not of Bak, whereas overexpression of Puma activated both (Shibue et al., 2006). This observation is supported by studies analysing mitochondrial targeting of Noxa in isolated mitochondria of mouse and rat liver cells where Noxa-mediated cytochrome c release was reported to occur independent from Bak oligomerization (Seo et al., 2003). Taken together, these observations suggest that Noxa kills cells in a mainly Bax-dependent but Bak-independent manner. However, this is somewhat surprising, given that Noxa has been repeatedly implicated in the disruption of Mcl1/Bak complexes, triggering Bak oligomerization during apoptosis induction in cells as diverse as MEF, HEK-293T, multiple myeloma and B-cell lymphomas (Willis et al., 2005; Inoue et al., 2007; Morales et al., 2008). This apparent discrepancy might be explained by the fact that oncogenic stress may not only impact expression of prosurvival molecules but also their subcellular localization as repeatedly observed for Bax, which is usually found in the cytoplasm of primary cells, but redistributes to mitochondria after transformation. Once at the mitochondria, Bax may be restrained efficiently by Mcl1, and Mcl1/Bax complexes may then be targeted by Noxa. However, it remains unclear why Noxa should selectively trigger oligomerization of Bax under these conditions, unless Mcl1 binds much more avidly to Bax than Bak, which may then be restrained by other prosurvival molecules such as Bclxl (Willis et al., 2005). Whatever the molecular mechanism may be, it is interesting to note that although primary MEF engage both Noxa and Puma for cell killing in response to UV irradiation, in E1a/ras-transformed cells Puma becomes dispensable for this process and Noxa accounts for all p53-induced death observed (Naik et al., 2007).

In summary, these data indicate that Noxa can engage Bax or Bak for apoptosis induction, but the engagement of these proteins depends on the cell type, cellular state and apoptotic stimulus applied. Furthermore, Noxa-induced cell death may become more relevant for apoptosis induction in malignant or highly proliferating versus primary or differentiated cells, suggesting its importance as tumor suppressor with therapeutic potential (see below).

Regulation of Mcl1 stability by Noxa

Given the fact that Noxa exclusively binds to Mcl1 and Bfl1/A1, cellular levels of these proteins determine susceptibility to Noxa-induced cell death of a given cell type. Inhibition or elimination of Mcl1 in response to cytotoxic signals is considered critical in cell death in a number of normal as well as malignant cells (Craig, 2002; Willis et al., 2005). However, targeting Mcl1 by transgenic overexpression of Noxa failed to induce significant cell death, for example, in immortalized MEF or NIH3T3 fibroblasts (Shibue et al., 2006), where survival depends on two or more Bcl2-like proteins such as Mcl1 and Bclxl (Willis et al., 2005). Consistently, Noxa needs to be functionally complemented by Bad, which targets Bcl2, Bclxl and Bclw, and overexpression of both BH3-only proteins induced cell death as efficiently as transgenic expression of Bim or Puma, able to neutralize all prosurvival Bcl2-like molecules (Chen et al., 2005).

An intriguing feature of Noxa is that it targets Mcl1 for proteasomal degradation. This event is considered to be a prerequisite for cell death in response to UV irradiation (Nijhawan et al., 2003), cytokine deprivation (Opferman et al., 2003), treatment with anticancer agents such as arsenic trioxide (Morales et al., 2008) or histone deacetylase inhibitors (Inoue et al., 2007). Although basal levels of Mcl1 appear to be regulated by the HECT- and BH3-domain-containing E3-ligase Mule/ARF-BP1 (Zhong et al., 2005), it is still unclear what regulates its degradation when complexed with Noxa, because the binding pocket is occupied with its BH3 domain. Interestingly, degradation of Mcl1 is not, as initially assumed, a prerequisite for apoptosis to occur. Interaction with Bim, for example, in response to GC treatment of acute lymphoblastic juvenile leukemia cells (Ploner et al., 2008), Puma overexpression (Mei et al., 2005) or arsenic trioxide treatment of myeloma cells (Morales et al., 2008), actually leads to its stabilization, presumably due to exclusion of Mule/ARF-BP1. The signature for degradation of the Mcl1/Noxa complex appears to be encoded in the C-terminal portion of the BH3 domain of Noxa (FRQKLL in human Noxa), as replacement of these residues with aa residues found in the same position in Bim (AYYARR) led to Mcl1 stabilization (Czabotar et al., 2007). Interestingly, although the Noxa BH3 domain in the context of Bim protein caused Mcl1 degradation, it failed to do so in the context of Bad, suggesting that residues outside the BH3 domain, functionally conserved in Noxa and Bim, contribute to this process (Czabotar et al., 2007).

Together all these studies reveal that Mcl1 degradation during cell death is uniquely associated with the formation of the Mcl1/Noxa complex whereas Mcl1/Bim or Mcl1/Puma interaction leads to its stabilization. This is of relevance considering the design of BH3 mimetics that may be able to bind to Mcl1, but may not necessarily promote its degradation. The selective (dis)advantage of Mcl1 degradation over stabilization during apoptosis induction, however, remains unclear at present. Identification of an Mcl1/Noxa-specific E3-ligase may allow some insight and rational explanation.

Although protein stability of the prosurvival Bcl2 homologue Bfl1/A1 also appears to be regulated by the proteasome and stability can be increased by Bim binding (Morales et al., 2008), no data are available addressing the possibility that A1/Noxa interactions may promote its degradation during apoptosis induction.

Possible physiological functions for Noxa

Noxa and the DNA-damage response

Two laboratories generated Noxa−/− mice to study its function in vivo (Shibue et al., 2003; Villunger et al., 2003). Mice lacking Noxa show no overt phenotype, but more detailed analysis of stress responses confirmed a function for Noxa in p53-dependent, but also p53-independent cell death after DNA damage, albeit in a cell type and stimulus-dependent manner. Although hemopoietic cells from Puma−/− mice resisted cell death induced by DNA damage or factor deprivation, cells from Noxa−/− mice were normally sensitive to cell death induction. However, loss of either protein protected MEF cells from DNA-damage-induced apoptosis caused by etoposide treatment, subsequently also confirmed in Noxa−/−Puma−/− MEF that showed significantly greater resistance as cells from single knockout mice. Analysis of Noxa−/−Puma−/− mice revealed a major nonredundant function for Puma in apoptosis induced by γ-irradiation in MEF and most lymphocyte subsets, besides CD4+8+ thymocytes, where Puma and Noxa account for all cell death induced by p53. Similar observations were made in etoposide-treated MEF transformed with E1a (Michalak et al., 2008). Interestingly, as mentioned before, Noxa and Puma contribute equally to apoptosis induced by UV irradiation in primary MEF, but Noxa becomes the dominant cell death inducer in response to this stimulus after oncogenic transformation (Naik et al., 2007). Noteworthy, this pathway to apoptosis depends on c-Jun N-terminal kinase activation (Tournier et al., 2000) and Mcl1 degradation (Nijhawan et al., 2003) that is triggered selectively by binding of Noxa to Mcl1.

Shibue et al. examined the involvement of Noxa in the DNA damage response in vivo by analysing apoptosis of epithelial cells in the stem cell region of small intestinal crypts and cells in lamina propria after whole-body γ-irradiation. Noxa deficiency protected both cell types from cell death. This is of particular interest because cells in lamina propria still die in the absence of p53 (Shibue et al., 2003). Protection from gastrointestinal apoptosis by loss of Noxa also led to a delayed onset of γ-irradiation disease-associated mortality. Noxa−/− mice survived significantly longer than wt or p53−/− animals. Taken together these results suggest that Noxa can be activated in response to γ-irradiation in a p53-dependent as well as -independent manner and may be prominent in the regulation of stem cell homeostasis, at least after DNA damage, as recently suggested also for Puma (see Review by Yu and Zhang, in this issue on page S71).

Possible functions for Noxa in the immune system

Although initial analysis of Noxa−/− mice did not reveal any abnormalities in the hematopoietic system, Noxa appears to control maintenance of memory CD4+ T Th1/Th2 cell homeostasis. As discussed in Chapter 2, Yamashita et al. (2008) reported that the excessive Th2 memory T-cell death observed in mice defective in the PcG gene Bmi1 was at least in part due to lacking repression of Noxa gene expression by Bmi1. In support, ablation of Noxa expression on a Bmi1 hemizygous background rescued the observed defects in memory cell generation, consistent with a key role for Noxa in the maintenance of Th2 memory T-cell homeostasis.

Interestingly, in contrast to memory Th2 cells, induction of Noxa per se was inefficient in triggering significant cell death in proliferating T cells, presumably due to concomitant induction of prosurvival Bcl2-like molecules. It may, however, prime proliferating T cells for death when conditions no longer favor clonal expansion, for example, when growth factors or cytokines become limiting at the end of an immune response. This idea is also supported by the observations that Noxa is induced by the common γ-chain of the receptors for cytokines IL-7 and IL-15, able to promote cell division in the absence of antigenic stimulation in CD8+ T cells. However, loss of Noxa mildly interferes with IL-2-deprivation-induced death of activated T cells, albeit not as efficiently as loss of Bim or Puma (Bauer et al., 2006), as well as IL-15-deprivation-induced death of NK-cells, together with Bim (Huntington et al., 2007). Consistent with its role as a sentinel in proliferating cells, Jurkat T-cell clones carrying shRNAs-targeting Noxa showed better survival under conditions of metabolic stress due to glucose limitation. Subsequent biochemical analysis indicated that the high Noxa levels induced during the proliferative response contributed to cell death by neutralizing the prosurvival effects of Mcl1 and presumably also Bfl1/A1 (Alves et al., 2006). Therefore, we speculate that upregulation of Noxa in proliferating T cells might counterbalance enhanced expression of prosurvival molecules, thereby helping to restrain oncogenic transformation and/or autoimmunity. However, because Noxa-deficient mice develop neither of these pathologies, Noxa may perform these functions only in conjunction with other cell death regulators and/or control mechanisms.

Noxa in tumor development

Lessons from mice and humans

Mice lacking both or even only a single allele of p53 develop tumors with high incidence. In contrast, no spontaneous tumor development was observed in Noxa−/− and even Noxa−/−Puma−/− double-deficient animals, up to an observation period of more than 1 year (Shibue et al., 2003; Michalak et al., 2008). This confirms that inactivation of the proapoptotic function of p53 is insufficient to promote tumor development but that two or more of its activities, such as induction of cell-cycle arrest and apoptosis, have to be impaired simultaneously. Interestingly, studies with knock-in mice that express an apoptosis-defective, but cell-cycle inhibitory competent mutant of p53 (R172P) indicate an equally prominent role for these two p53 functions in the suppression of thymic lymphoma development (Liu et al., 2004). Whether loss of Noxa may impact tumor formation caused by DNA damage remains to be tested and it will be interesting to see if loss of Noxa on a p21-deficient background increases the rate of spontaneous tumorigenesis in animal lacking this CDK inhibitor.

So far, the connection between Noxa and tumor suppression is mainly circumstantial. In mice bearing a liver-specific deletion of c-Jun, the development of chemically induced hepatocellular carcinoma (HCC) is significantly delayed, revealing the oncogenic potential of c-Jun in an in vivo model (Eferl et al., 2003). c-Jun, a central component of the transcription factor Ap1, is a major regulator of proliferation and apoptosis in liver cells and loss of c-Jun in HCC leads to an increased number of apoptotic cells that display increased mRNA levels of p53 and Noxa (Eferl et al., 2003). Induction of Noxa in the absence of c-Jun seems to be specific, because the expression of other proapoptotic p53 target genes, such as Bax, Puma or Apaf-1, was unaffected. However, it was not formally demonstrated that the observed upregulation of Noxa mRNA was the consequence of p53 activation, leaving the possibility that Noxa and p53 may be independently activated by loss of c-Jun. Taken together, these observations indicate that c-Jun promotes tumor cell survival by inhibiting the proapoptotic action of p53, maybe by inhibiting expression of Noxa.

Most recently, the impact of loss of Noxa on c-Myc-driven B-cell lymphomagenesis has been studied. Aberrant expression of c-Myc leads to p53 activation and subsequent induction of apoptosis that may engage Noxa and/or Puma to restrain tumor formation. -Myc transgenic mice lacking Puma develop lymphomas significantly earlier than Eμ-Myc mice, consistent with the initial hypothesis (Hemann et al., 2004; Garrison et al., 2008). Loss of Noxa, however, did not accelerate tumor formation, but only did so on a sensitized background, (haplo)insufficient for Puma (Michalak et al., 2009). This observation suggests a modifier, rather than a tumor suppressor function for Noxa, at least in this model system. Even more puzzling, loss of Noxa clearly delayed the onset of c-Myc-driven immature preB cell, but had no impact on the development of mature IgM+ B-cell tumors. Although it is unclear whether Noxa is induced on c-Myc-driven oncogenic stress in IgM+ B cells, one may speculate that in its absence compensatory activation of other BH3-only proteins such as Puma or Bim may be more effective in removing (pre)malignant preB cells. Although this explanation remains to be formerly tested, the finding itself suggests that under certain conditions BH3-only proteins possess oncogenic potential (Michalak et al., 2008).

Following up on the idea that tumors with functional p53 may have lost Noxa function, Lee et al. (2003) investigated 78 colon adenocarcinomas, 53 advanced gastric adenocarcinomas, 83 non-small-cell lung cancers, 76 breast carcinomas, 33 urinary bladder transitional cell carcinomas and 90 HCCs for their Noxa status. However, apart from one transitional cell carcinoma of the urinary bladder, which carried a somatic mutation in the Noxa gene, all tumor samples contained wt Noxa sequences. Moreover, when analysed in vitro the observed mutation had no impact on the proapoptotic activity of Noxa (Lee et al., 2003). Comparison of Noxa mRNA expression levels in 94 colorectal adenocarcinoma and adjacent normal mucosa samples by quantitative RT-PCR failed to reveal any pathology-associated differences, nor were mutations found in the Noxa coding sequence (Jansson et al., 2003). Interestingly, though, integrative genomic analysis of small-cell lung carcinoma revealed a correlation between amplification on 18q and sensitivity to the BH3-mimetic ABT-737 (Olejniczak et al., 2007). Resistance of cancer cells to ABT-737 is strongly associated with the relative expression of Mcl1, which it cannot antagonize (see below and review by Labi et al., 2008). The region of gain contains Bcl2 that can be potently blocked by ABT-737, but also Noxa that effectively neutralizes Mcl1 (van Delft et al., 2006), suggesting that 18q21–23, or maybe even Noxa expression alone, may be a clinically relevant predictor for responsiveness to Bcl2 family inhibitors (Olejniczak et al., 2007). Along that line, in B-CLL cells expression of Noxa protein was found frequently increased, when compared with tonsillar B cells (Mackus et al., 2005) and may be causal for the high sensitivity of these tumors to ABT-737 treatment (Oltersdorf et al., 2005).

Noxa expression levels, however, failed to correlate with overall or disease-free survival in human colon carcinoma, whereas relative expression levels of Bim or Puma did correlate well with these parameters (Sinicrope et al., 2008). Finally, Noxa was also found deleted in the Burkitt-derived cell line Elijah and loss of one allele was observed in the related Karpas 106, UPN1 and Namalwa cell lines (Mestre-Escorihuela et al., 2007). Two of these cell lines showed inactivating mutations in the remaining allele. Mutational screening in 15 primary B-NHL biopsies, however, failed to reveal inactivating mutations but identified polymorphisms in 3 out of 15 promoter regions analysed. Analysis of protein expression by immunohistochemistry on biopsy samples from 367 B-NHL patients failed to reveal significant disease-associated changes, but resembled the expression found in the nonmalignant counterpart of these tumor cells (Mestre-Escorihuela et al., 2007), suggesting that deregulation of Noxa expression is not significant in the pathogenesis of B-NHL.

Noxa and the cellular response to anticancer agents

Although a possible involvement of Noxa in cancerogenesis is still under investigation, its importance as a drug target became evident when analysing the efficiency of the BH3-mimetic ABT-737. Using a nutation nuclear magnetic resonance structure-based approach to target the BH3-binding groove of Bclxl, Abbott Laboratories designed a cell permeating BH3 mimetic (Oltersdorf et al., 2005) that efficiently antagonized Bcl2, Bclxl and Bclw, but was ineffective in killing tumor cells expressing high levels of Mcl1 (van Delft et al., 2006; Tahir et al., 2007; Konopleva et al., 2008). Knockdown of Mcl1 helped to restore sensitivity to ABT-737 treatment in per se resistant colon and bladder cell lines (Lin et al., 2007). So, to overcome ABT resistance development of a Noxa-like BH3 mimetic or a mimetic that also targets Mcl1 is highly desirable (reviewed in more detail by Letai and Ni Chonghaile, in this issue on page S149).

As indicated Noxa is not only clearly relevant for p53-induced apoptosis triggered by classical anticancer agents such as etoposide or γ-irradiation but also for tumor cell death triggered by the cyclin-dependent kinase inhibitor R-roscovitine, actually leading to reduced Mcl1 protein levels (Hallaert et al., 2007), inhibition of histone deacetylases, triggering de novo synthesis of Noxa (Inoue et al., 2007) or, somewhat counterintuitive, proteasome inhibition. In malignant melanoma, but not primary melanocytes, as well as multiple myeloma, but not peripheral blood lymphocytes, nonselective inhibition of the proteasome by Bortezomib/Velcade causes an increase in the expression of Noxa preceding tumor cell apoptosis. In contrast, Bim is induced by this treatment in normal as well in cancer cells, but the former are highly resistant to this drug (Fernandez et al., 2005; Qin et al., 2005). Noteworthy, induction of Noxa is not simply a consequence of impaired proteasomal degradation of Noxa/Mcl1 complexes but occurs also at the transcriptional level in a c-Myc-dependent manner and independent from the p53 status of the tumor cell (Nikiforov et al., 2007). For a more detailed review on this topic see the chapter by M Soengas (in this issue). Taken together, a correlation between proliferation potential of the cell and sensitivity toward Noxa becomes again apparent, suggesting a possible link between the formation of Mcl1/Noxa complexes, cell-cycle progression and apoptosis induction.

Conclusions

Although so far only a limited correlation between the expression of Noxa and tumor development has been found in humans, activation of this BH3-only protein appears critical for the cellular response to anticancer treatment regimens, such as γ-irradiation, the application of chemotherapeutic drugs, and also novel compounds such as the BH3-mimetic ABT-737. Restoring or enhancing the tumor cell ability to increase Noxa for targeting Mcl1 and/or Bfl1/A1 expression or mimicking this function by small molecular compounds should significantly increase treatment efficiency of current and novel treatment strategies against cancer. However, a more detailed knowledge on Noxa biology is required, because aberrant activation of this BH3-only protein may well compromise the efficiency of the immune system, for example, by prompting memory T-cell death, or by limiting the clonal expansion of antigen-activated T cells.

Acknowledgements

The work in our laboratories was supported by fellowships and grants from the Austrian Science Fund (FWF): Y212-B13 START, the Doctoral College MCBO, the SFB021, projects P18747 and P18571, the Association for International Cancer Research (AICR), EU-FP7 (ApopTrain) and the Tyrolean Science Fund (TWF). We apologize to the many scientists in this field whose excellent research was not cited but was only referred to indirectly through reviews.

Footnotes

Conflict of interest

The authors declare no conflict of interest.

References

  1. Alves NL, Derks IA, Berk E, Spijker R, van Lier RA, Eldering E. The Noxa/Mcl-1 axis regulates susceptibility to apoptosis under glucose limitation in dividing T cells. Immunity. 2006;24:703–716. doi: 10.1016/j.immuni.2006.03.018. [DOI] [PubMed] [Google Scholar]
  2. Bauer A, Villunger A, Labi V, Fischer SF, Strasser A, Wagner H, et al. The NF-kappaB regulator Bcl-3 and the BH3-only proteins Bim and Puma control the death of activated T cells. Proc Natl Acad Sci USA. 2006;103:10979–10984. doi: 10.1073/pnas.0603625103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG, et al. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 2005;17:393–403. doi: 10.1016/j.molcel.2004.12.030. [DOI] [PubMed] [Google Scholar]
  4. Coultas L, Huang DC, Adams JM, Strasser A. Pro-apoptotic BH3-only Bcl-2 family members in vertebrate model organisms suitable for genetic experimentation. Cell Death Differ. 2002;9:1163–1166. doi: 10.1038/sj.cdd.4401096. [DOI] [PubMed] [Google Scholar]
  5. Craig RW. MCL1 provides a window on the role of the BCL2 family in cell proliferation, differentiation and tumorigenesis. Leukemia. 2002;16:444–454. doi: 10.1038/sj.leu.2402416. [DOI] [PubMed] [Google Scholar]
  6. Czabotar PE, Lee EF, van Delft MF, Day CL, Smith BJ, Huang DC, et al. Structural insights into the degradation of Mcl-1 induced by BH3 domains. Proc Natl Acad Sci USA. 2007;104:6217–6222. doi: 10.1073/pnas.0701297104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Eferl R, Ricci R, Kenner L, Zenz R, David JP, Rath M, et al. Liver tumor development. c-Jun antagonizes the proapoptotic activity of p53. Cell. 2003;112:181–192. doi: 10.1016/s0092-8674(03)00042-4. [DOI] [PubMed] [Google Scholar]
  8. Fei P, Bernhard EJ, El-Deiry WS. Tissue-specific induction of p53 targets in vivo. Cancer Res. 2002;62:7316–7327. [PubMed] [Google Scholar]
  9. Fernandez Y, Verhaegen M, Miller TP, Rush JL, Steiner P, Opipari AW, et al. Differential regulation of noxa in normal melanocytes and melanoma cells by proteasome inhibition: therapeutic implications. Cancer Res. 2005;65:6294–6304. doi: 10.1158/0008-5472.CAN-05-0686. [DOI] [PubMed] [Google Scholar]
  10. Flinterman M, Guelen L, Ezzati-Nik S, Killick R, Melino G, Tominaga K, et al. E1A activates transcription of p73 and Noxa to induce apoptosis. J Biol Chem. 2005;280:5945–5959. doi: 10.1074/jbc.M406661200. [DOI] [PubMed] [Google Scholar]
  11. Garrison SP, Jeffers JR, Yang C, Nilsson JA, Hall MA, Rehg JE, et al. Selection against Puma gene expression in Myc-driven B-cell lymphomagenesis. Mol Cell Biol. 2008;28:5391–5402. doi: 10.1128/MCB.00907-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Hallaert DY, Spijker R, Jak M, Derks IA, Alves NL, Wensveen FM, et al. Crosstalk among Bcl-2 family members in B-CLL: seliciclib acts via the Mcl-1/Noxa axis and gradual exhaustion of Bcl-2 protection. Cell Death Differ. 2007;14:1958–1967. doi: 10.1038/sj.cdd.4402211. [DOI] [PubMed] [Google Scholar]
  13. Hemann MT, Zilfou JT, Zhao Z, Burgess DJ, Hannon GJ, Lowe SW. Suppression of tumorigenesis by the p53 target Puma. Proc Natl Acad Sci USA. 2004;101:9333–9338. doi: 10.1073/pnas.0403286101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Hershko T, Ginsberg D. Up-regulation of Bcl-2 homology 3 (BH3)-only proteins by E2F1 mediates apoptosis. J Biol Chem. 2004;279:8627–8634. doi: 10.1074/jbc.M312866200. [DOI] [PubMed] [Google Scholar]
  15. Hijikata M, Kato N, Sato T, Kagami Y, Shimotohno K. Molecular cloning and characterization of a cDNA for a novel phorbol-12-myristate-13-acetate-responsive gene that is highly expressed in an adult T-cell leukemia cell line. J Virol. 1990;64:4632–4639. doi: 10.1128/jvi.64.10.4632-4639.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Huntington ND, Puthalakath H, Gunn P, Naik E, Michalak EM, Smyth MJ, et al. Interleukin 15-mediated survival of natural killer cells is determined by interactions among Bim, Noxa and Mcl-1. Nat Immunol. 2007;8:856–863. doi: 10.1038/ni1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Inoue S, Riley J, Gant TW, Dyer MJ, Cohen GM. Apoptosis induced by histone deacetylase inhibitors in leukemic cells is mediated by Bim and Noxa. Leukemia. 2007;21:1773–1782. doi: 10.1038/sj.leu.2404760. [DOI] [PubMed] [Google Scholar]
  18. Jansson AK, Emterling AM, Arbman G, Sun XF. Noxa in colorectal cancer: a study on DNA, mRNA and protein expression. Oncogene. 2003;22:4675–4678. doi: 10.1038/sj.onc.1206655. [DOI] [PubMed] [Google Scholar]
  19. Kim JY, Ahn HJ, Ryu JH, Suk K, Park JH. BH3-only protein Noxa is a mediator of hypoxic cell death induced by hypoxia-inducible factor 1alpha. J Exp Med. 2004;199:113–124. doi: 10.1084/jem.20030613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Konopleva M, Watt J, Contractor R, Tsao T, Harris D, Estrov Z, et al. Mechanisms of antileukemic activity of the novel Bcl-2 homology domain-3 mimetic GX15-070 (Obatoclax) Cancer Res. 2008;68:3413–3420. doi: 10.1158/0008-5472.CAN-07-1919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Labi V, Grespi F, Baumgartner F, Villunger A. Targeting the Bcl-2-regulated apoptosis pathway by BH3 mimetics: a break-through in anticancer therapy? Cell Death Differ. 2008;15:977–987. doi: 10.1038/cdd.2008.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Lau LM, Nugent JK, Zhao X, Irwin MS. HDM2 antagonist Nutlin-3 disrupts p73-HDM2 binding and enhances p73 function. Oncogene. 2008;27:997–1003. doi: 10.1038/sj.onc.1210707. [DOI] [PubMed] [Google Scholar]
  23. Lee SH, Soung YH, Lee JW, Kim HS, Lee JH, Kim HS, et al. Mutational analysis of Noxa gene in human cancers. APMIS. 2003;111:599–604. doi: 10.1034/j.1600-0463.2003.1110602.x. [DOI] [PubMed] [Google Scholar]
  24. Lin X, Morgan-Lappe S, Huang X, Li L, Zakula DM, Vernetti LA, et al. ‘Seed’ analysis of off-target siRNAs reveals an essential role of Mcl-1 in resistance to the small-molecule Bcl-2/Bcl-XL inhibitor ABT-737. Oncogene. 2007;26:3972–3979. doi: 10.1038/sj.onc.1210166. [DOI] [PubMed] [Google Scholar]
  25. Liu G, Parant JM, Lang G, Chau P, Chavez-Reyes A, El-Naggar AK, et al. Chromosome stability, in the absence of apoptosis, is critical for suppression of tumorigenesis in Trp53 mutant mice. Nat Genet. 2004;36:63–68. doi: 10.1038/ng1282. [DOI] [PubMed] [Google Scholar]
  26. Liu X, Dai S, Zhu Y, Marrack P, Kappler JW. The structure of a Bcl-xL/Bim fragment complex: implications for Bim function. Immunity. 2003;19:341–352. doi: 10.1016/s1074-7613(03)00234-6. [DOI] [PubMed] [Google Scholar]
  27. Mackus WJ, Kater AP, Grummels A, Evers LM, Hooijbrink B, Kramer MH, et al. Chronic lymphocytic leukemia cells display p53-dependent drug-induced Puma upregulation. Leukemia. 2005;19:427–434. doi: 10.1038/sj.leu.2403623. [DOI] [PubMed] [Google Scholar]
  28. Mei Y, Du W, Yang Y, Wu M. Puma(*)Mcl-1 interaction is not sufficient to prevent rapid degradation of Mcl-1. Oncogene. 2005;24:7224–7237. doi: 10.1038/sj.onc.1208873. [DOI] [PubMed] [Google Scholar]
  29. Mestre-Escorihuela C, Rubio-Moscardo F, Richter JA, Siebert R, Climent J, Fresquet V, et al. Homozygous deletions localize novel tumor suppressor genes in B-cell lymphomas. Blood. 2007;109:271–280. doi: 10.1182/blood-2006-06-026500. [DOI] [PubMed] [Google Scholar]
  30. Michalak EM, Jansen ES, Happo L, Cragg MS, Tai L, Smyth GK, et al. Puma and to a lesser extent Noxa are suppressors of Myc-induced lymphomagenesis. Cell Death Differ. 2009 doi: 10.1038/cdd.2008.195. e-pub ahead of print 16 January 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Michalak EM, Villunger A, Adams JM, Strasser A. In several cell types tumour suppressor p53 induces apoptosis largely via Puma but Noxa can contribute. Cell Death Differ. 2008;15:1019–1029. doi: 10.1038/cdd.2008.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Morales AA, Gutman D, Lee KP, Boise LH. BH3-only proteins Noxa, Bmf, and Bim are necessary for arsenic trioxide-induced cell death in myeloma. Blood. 2008;111:5152–5162. doi: 10.1182/blood-2007-10-116889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Naik E, Michalak EM, Villunger A, Adams JM, Strasser A. Ultraviolet radiation triggers apoptosis of fibroblasts and skin keratinocytes mainly via the BH3-only protein Noxa. J Cell Biol. 2007;176:415–424. doi: 10.1083/jcb.200608070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Nijhawan D, Fang M, Traer E, Zhong Q, Gao W, Du F, et al. Elimination of Mcl-1 is required for the initiation of apoptosis following ultraviolet irradiation. Genes Dev. 2003;17:1475–1486. doi: 10.1101/gad.1093903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Nikiforov MA, Riblett M, Tang WH, Gratchouck V, Zhuang D, Fernandez Y, et al. Tumor cell-selective regulation of NOXA by c-MYC in response to proteasome inhibition. Proc Natl Acad Sci USA. 2007;104:19488–19493. doi: 10.1073/pnas.0708380104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T, Yamashita T, et al. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science. 2000;288:1053–1058. doi: 10.1126/science.288.5468.1053. [DOI] [PubMed] [Google Scholar]
  37. Olejniczak ET, Van SC, Anderson MG, Wang G, Tahir SK, Sauter G, et al. Integrative genomic analysis of small-cell lung carcinoma reveals correlates of sensitivity to bcl-2 antagonists and uncovers novel chromosomal gains. Mol Cancer Res. 2007;5:331–339. doi: 10.1158/1541-7786.MCR-06-0367. [DOI] [PubMed] [Google Scholar]
  38. Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005;435:677–681. doi: 10.1038/nature03579. [DOI] [PubMed] [Google Scholar]
  39. Opferman JT, Letai A, Beard C, Sorcinelli MD, Ong CC, Korsmeyer SJ. Development and maintenance of B and T lymphocytes requires antiapoptotic MCL-1. Nature. 2003;426:671–676. doi: 10.1038/nature02067. [DOI] [PubMed] [Google Scholar]
  40. Ploner C, Rainer J, Niederegger H, Eduardoff M, Villunger A, Geley S, et al. The BCL2 rheostat in glucocorticoid-induced apoptosis of acute lymphoblastic leukemia. Leukemia. 2008;22:370–377. doi: 10.1038/sj.leu.2405039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Qin JZ, Ziffra J, Stennett L, Bodner B, Bonish BK, Chaturvedi V, et al. Proteasome inhibitors trigger NOXA-mediated apoptosis in melanoma and myeloma cells. Cancer Res. 2005;65:6282–6293. doi: 10.1158/0008-5472.CAN-05-0676. [DOI] [PubMed] [Google Scholar]
  42. Schmidt S, Rainer J, Riml S, Ploner C, Jesacher S,C, et al. Identification of glucocorticoid-response genes in children with acute lymphoblastic leukemia. Blood. 2006;107:2061–2069. doi: 10.1182/blood-2005-07-2853. [DOI] [PubMed] [Google Scholar]
  43. Schuler M, Maurer U, Goldstein JC, Breitenbucher F, Hoffarth S, Waterhouse NJ, et al. p53 triggers apoptosis in oncogene-expressing fibroblasts by the induction of Noxa and mitochondrial Bax translocation. Cell Death Differ. 2003;10:451–460. doi: 10.1038/sj.cdd.4401180. [DOI] [PubMed] [Google Scholar]
  44. Seo YW, Shin JN, Ko KH, Cha JH, Park JY, Lee BR, et al. The molecular mechanism of Noxa-induced mitochondrial dysfunction in p53-mediated cell death. J Biol Chem. 2003;278:48292–48299. doi: 10.1074/jbc.M308785200. [DOI] [PubMed] [Google Scholar]
  45. Shibue T, Suzuki S, Okamoto H, Yoshida H, Ohba Y, Takaoka A, et al. Differential contribution of Puma and Noxa in dual regulation of p53-mediated apoptotic pathways. EMBO J. 2006;25:4952–4962. doi: 10.1038/sj.emboj.7601359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Shibue T, Takeda K, Oda E, Tanaka H, Murasawa H, Takaoka A, et al. Integral role of Noxa in p53-mediated apoptotic response. Genes Dev. 2003;17:2233–2238. doi: 10.1101/gad.1103603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Sinicrope FA, Rego RL, Okumura K, Foster NR, O’Connell MJ, Sargent DJ, et al. Prognostic impact of bim, Puma, and noxa expression in human colon carcinomas. Clin Cancer Res. 2008;14:5810–5818. doi: 10.1158/1078-0432.CCR-07-5202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Tahir SK, Yang X, Anderson MG, Morgan-Lappe SE, Sarthy AV, Chen J, et al. Influence of Bcl-2 family members on the cellular response of small-cell lung cancer cell lines to ABT-737. Cancer Res. 2007;67:1176–1183. doi: 10.1158/0008-5472.CAN-06-2203. [DOI] [PubMed] [Google Scholar]
  49. Tournier C, Hess P, Yang DD, Xu J, Turner TK, Nimnual A, et al. Requirement of JNK for stress-induced activation of the cytochrome c-mediated death pathway. Science. 2000;288:870–874. doi: 10.1126/science.288.5467.870. [DOI] [PubMed] [Google Scholar]
  50. van Delft MF, Wei AH, Mason KD, Vandenberg CJ, Chen L, Czabotar PE, et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/ Bax if Mcl-1 is neutralized. Cancer Cell. 2006;10:389–399. doi: 10.1016/j.ccr.2006.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Villunger A, Michalak EM, Coultas L, Mullauer F, Bock G, Ausserlechner MJ, et al. p53- and drug-induced apoptotic responses mediated by BH3-only proteins Puma and noxa. Science. 2003;302:1036–1038. doi: 10.1126/science.1090072. [DOI] [PubMed] [Google Scholar]
  52. Wang H, Wang L, Erdjument-Bromage H, Vidal M, Tempst P, Jones RS, et al. Role of histone H2A ubiquitination in polycomb silencing. Nature. 2004;431:873–878. doi: 10.1038/nature02985. [DOI] [PubMed] [Google Scholar]
  53. Wang Z, Sun Y. Identification and characterization of two splicing variants of human Noxa. Anticancer Res. 2008;28:1667–1674. [PubMed] [Google Scholar]
  54. Willis SN, Chen L, Dewson G, Wei A, Naik E, Fletcher JI, et al. Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins. Genes Dev. 2005;19:1294–1305. doi: 10.1101/gad.1304105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Yamashita M, Kuwahara M, Suzuki A, Hirahara K, Shinnaksu R, Hosokawa H, et al. Bmi1 regulates memory CD4T cell survival via repression of the Noxa gene. J Exp Med. 2008;205:1109–1120. doi: 10.1084/jem.20072000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Zhong Q, Gao W, Du F, Wang X. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell. 2005;121:1085–1095. doi: 10.1016/j.cell.2005.06.009. [DOI] [PubMed] [Google Scholar]