Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

Cell, Tumor, and Stem Cell Biology

Proline Oxidase Functions as a Mitochondrial Tumor Suppressor in Human Cancers

Yongmin Liu,1 Gregory L. Borchert,1 Steven P. Donald,3 Bhalchandra A. Diwan,1 Miriam Anver,2 and James M. Phang3

1Basic Science Program and 2Pathology/Histotechnology Laboratory, Science Applications International Corporation-Frederick, Inc., and 3Laboratory of Comparative Carcinogenesis, Center for Cancer Research, National Cancer Institute, Frederick, Maryland

Abstract is to catalyze the rate-limiting two-electron oxidation of proline to D1-pyrroline-5-carboxylate (P5C) and the subsequent transfer Tumor metabolism and bioenergetics have become important topics for cancer research and are promising targets for of reducing equivalents from the reduced flavin cofactor to the anticancer therapy. Although glucose serves as the main mitochondrial electron transport chain (6). Together with P5C source of energy, proline, an alternative substrate, is reductase, which converts P5C backto proline, POX mediates the important, especially during nutrient stress. Proline oxidase proline cycle to shuttle redox equivalents between mitochondria (POX), catalyzing the first step in proline catabolism, is and cytosol. P5C is the intermediate providing a direct carbon a induced by p53 and can regulate cell survival as well as bridge connecting the TCA cycle through glutamate and - a mediate programmed cell death. In a mouse xenograft tumor ketoglutarate ( -KG) and to the urea cycle through ornithine (7). model, we found that POX greatly reduced tumor formation by Another important feature about proline is that together with causing G cell cycle arrest. Furthermore, immunohistochem- hydroxyproline, it constitutes over 25% of incorporated residues of 2 collagen, which is the most abundant in the extracellular ical staining showed decreased POX expression in tumor tissues. Importantly, HIF-1A signaling was impaired with POX matrix and, in fact, in the entire human body. When degraded expression due to the increased production of A-ketogluta- during inflammation and tumor progression, collagen releases a rate, a critical substrate for prolyl hydroxylation and large quantity of proline after digestion by matrix metalloprotei- degradation of HIF-1A. Combined with previous in vitro nases and prolidase. The contribution of these products to a variety findings and reported clinical genetic associations, these new of cellular processes has recently been recognized (8). findings lead us to propose POX as a mitochondrial tumor Recent research has revealed the response of POX to energy and suppressor and a potential target for cancer therapy. [Cancer nutrient stress. After it was found to be one of markedly Res 2009;69(16):6414–22] induced by p53 (9), its role in cell proliferation, apoptotic cell death, particularly in cancer cells has been intensively investigated. POX negatively regulates the growth of various types of cancer cells, Introduction which prompted us to investigate further its potential role in human cancer development. In this study, we used multiple Although controversial over the years, a molecular basis for the approaches to define the role of POX in tumor suppression and ‘‘Warburg effect’’ is emerging from genetic and pharmacologic characterized mediating mechanisms. In addition to inducing studies that show that specific oncogene and tumor suppressor apoptotic cell death, POX also caused cell cycle arrest at G phase. mutations directly regulate glycolysis, oxidative phosphorylation, 2 Furthermore, we found that POX inhibited tumor formation in a and other energy/nutrient supply processes (1–5). The combina- mouse xenograft model. Importantly, in human tissue samples, tion of these mutations and the hypoxic conditions in many tumor POX expression levels were much lower in tumor tissues than in types is likely to control synergistically the overall metabolic state their normal tissue counterparts. Finally, HIF signaling was reduced and the fate of individual tumors. There are several mitochondrial in POX-expressing cells. These findings, combined with published tumor suppressors that have been identified (5). The critical ways data and genetic information, suggest that POX is a mitochondrial they exert their antitumor effect are through hypoxia inducible tumor suppressor protein. factor (HIF), a key player in tumor formation, and affects the tricarboxylic acid (TCA) cycle. Our previous and current findings indicate proline oxidase (POX) plays a similar role and is a mitochondrial tumor suppressor. Materials and Methods POX, also called proline dehydrogenase, is a mitochondrial Cell culture. The DLD-1 human colon cancer cells are from America inner-membrane enzyme involved in the degradation of the Type Culture Collection. The generation, characterization, and maintenance secondary amino acid proline. The function of this flavoenzyme of the DLD-1 Tet-Off POX and DLD-1 Tet-Off Vector cell lines have been previously described (10, 11). Dimethyloxalylglycine was purchased from Frontier Scientific. Doxycycline (DOX) was purchased from Sigma. Note: Supplementary data for this article are available at Cancer Research Online Multiple protein sequence alignments. These sequence alignments (http://cancerres.aacrjournals.org/). were done using the CLUSTALW2 program at the Multiple Sequence Requests for reprints: Yongmin Liu, Basic Science Program, Science Applications Alignment Web site.4 The amino acid sequences were aligned and International Corporation-Frederick, Inc., Frederick and National Cancer Institute at compared among the species of Homo sapiens, Mus musculus, Rattus Frederick, Frederick, MD 21702. Phone: 301-846-5370; Fax: 301-402-0153; E-mail: [email protected] or James Phang, Laboratory of Comparative Carcinogenesis, norvegicus, Danio rerio, and Drosophila melanogaster. Center for Cancer Research, National Cancer Institute, Frederick, MD 21702. Phone: 301-846-5367; E-mail: [email protected]. I2009 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-09-1223 4 http://align.genome.jp/

Cancer Res 2009;69: (16). August 15, 2009 6414 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

POX: A Novel Mitochondrial Tumor Suppressor

Figure 1. In vivo tumor growth inhibition. DLD-1 Tet-off POX cells and vector control cells were injected into Balb/Cnude mice. A, numbers of mice that developed tumors after injection of cells when mice were given water with or without DOX. Left, mice injected with POX cells; right, mice injected with vector cells. For those mice injected with POX cells in the (À) DOX group that did not develop tumors, half (eight mice) were switched to receive water with DOX. Lower left, the number of mice developing tumors was recorded. Lower right, H&E staining and immunohistochemical staining for POX show the induced expression and location of POX. B, left, TUNEL assay showed relatively high apoptosis induction in POX-expressing cells. Right, quantitation of at least three slide preparations. Columns, mean; bars, SD. C, immunohistochemical staining shows the reduced BrdUrd incorporation by POX. Right, quantification from at least three slides. Columns, mean; bars, SD. D, left, soft agar assay showed reduced clonogenic ability of DLD-1 cells by POX. Right, quantitation of clones per well; represents data F of at least four determinations. *, statistically significant difference (*, P < 0.05; **, P < 0.01). Magnification for A to C was Â40 or Â100.

www.aacrjournals.org 6415 Cancer Res 2009;69: (16). August 15, 2009

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

Cancer Research

Cell growth assays. The CellTiter 96 aqueous nonradioactive cell Semiquantitative reverse transcription-PCR analysis. Total RNA was proliferation assay (MTS assay; Promega), performed according to the prepared from DLD-1-POX cells using Qiagen RNeasy kits (Qiagen). Reverse manufacturer’s protocol, was described in our previous studies (11). Each transcription-PCR (RT-PCR) reactions were performed using Ready-To-Go data point was performed in triplicate, and the results were reported as RT-PCR Beads (Amersham). The RT reaction was carried out using 0.5 Ag mean absorption F SE. RNA at 42jC for 30 min, followed by 5 min at 95jC to inactivate the Western blot analysis. The methods for cell lysate preparation, protein enzyme. Then, 2 Amol/L specific primers were added per reaction and concentration determination, and electrophoresis have been described subjected to 23 to 28 cycles of amplification at 94jC 30 s/60jC 30 s/72jC previously (11). The primary antibodies used were GADD45a (Chemicon) 45 s. RT-PCR to actin was performed as control. All PCR shown are GADDh, GADDg, GADD34, GADD153 (Santa Cruz Co.), HIF-1a, vascular representative of at least two independent experiments. The specific endothelial growth factor (VEGF), geminin, cyclin B1, p21, cdc25c, cdc2, sequences of the primers used have been reported elsewhere (12–15). p-cdc2 (tyr-15), p-cdc2 (thr-161; Cell Signaling), (Calbiochem), and actin Animal studies. The animal protocol was approved by the National (Sigma). The anti-POX polyclonal antibody was prepared and tested in the Cancer Institute-FrederickAnimal Care and Use committee. Sixty-two laboratory. Blots were developed using the chemiluminescence procedure Balb/C athymic mice were randomized to four groups. Two groups with (Amersham). All Western blots shown are representative of at least two nine mice in each were injected with 2 Â 106 of DLD-1 Tet-off Vector cells independent experiments. fed water with or without 100 Ag/mL of DOX. The other two groups, with Flow cytometry. The methods have been reported previously (11). DLD-1 22 mice in each, were given same number of DLD-1 Tet-off POX cells, also in Tet-Off POX cells were cultured in the medium with various concentrations (+) DOX or (À) DOX conditions. For mice that did not develop tumors in of DOX. Propidium iodide–stained cells were analyzed with an EPICS-XL- (À) DOX group, half of them (eight mice) were switched to receive drinking MCL flow cytometer (Beckman Coulter, Inc.). Each data point was water with DOX, whereas the other half were kept in (À) DOX. The tumor performed in triplicate, and the results were reported as the mean F SE. development in these two new groups was monitored. The mice were

Figure 2. POX caused G2 cell cycle arrest. A, left, POX was dose-dependently repressed by increasing DOX concentrations in DLD-1 Tet-Off POX cells. Right, cell growth assay showed increasing DOX concentrations released DLD-1 cells from POX-mediated inhibition of growth. B, flow cytometry assay showed that increasing POX expression with decreasing DOX concentration induced G2 cell cycle arrest. C, Western blot showed the induction of Geminin, by increasing POX expression with decreasing DOX. D, the effects of POX expression with decreasing DOX concentrations on several important regulators of G2 phase determined by Western blots.

Cancer Res 2009;69: (16). August 15, 2009 6416 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

POX: A Novel Mitochondrial Tumor Suppressor

Figure 3. The involvement of GADD family . The DLD-1 Tet-off POX cells were cultured in medium with 20 ng/mL [(+) DOX], or 100- and 1000-fold dilution (0.2 or 0.02 ng/mL) for 1 or 3 d. The cells were harvested and RNA and cell lysates were prepared and then semiquantitative RT-PCR (A and B) and Western blots (C) for GADD members were performed. Actin was used as loading control.

observed for tumor development thrice a weekand tumors were measured POX overexpression inhibited tumor development in a twice a weekafter they developed. mouse xenograft model. We and others have shown that POX Immunohistochemical staining. The immunostaining for POX was inhibited cell growth in a variety of cultured cancer cells (11, 18–21). performed by the Pathology/Histotechnology Laboratory, National Cancer To extend this cancer-suppressing effect to in vivo studies, we Institute-Frederick. The human tissue arrays, which contained paired developed a mouse xenograft model by injecting DLD-1 Tet-off cancer/normal tissues from the same patient, were purchased from Cybridi, Co. Two independent pathologists read and graded the expression as 5 POX cells and DLD-1 Tet-off Vector cells into athymic mice. In mice levels: 0, +, ++, +++, and ++++. The expression of POX in tumor and receiving vector control cells, tumors developed quickly. The normal tissues from the same patients were then compared, and changes addition of DOX to the drinking water made no difference for (up-regulation, no change, or down-regulation) were determined. Data tumor development (Fig. 1A). For mice injected with DLD-1-POX were statistically analyzed using the Z test. The xenograft tumors were cells, in the (+) DOX group, as was the case with vector controls, collected from athymic mice and fixed in 10% formaldehyde and then tumors developed quickly. By contrast, tumor development was embedded in paraffin. Tissue sections were then mounted on slides and greatly reduced in mice in the (À) DOX group (Fig. 1A). To extend processed for either H&E staining or immunostaining for POX, BrdUrd, or this experiment further, we switched half (8) of the mice in the (À) for terminal dUTP nick-end labeling (TUNEL) assay. DOX group that were tumor-free by week5 to receive drinking Determination of the A-keto acids. a-Keto acids were determined by water with DOX to suppress POX expression, and then compared high-performance liquid chromatography (HPLC) as their fluorescent quinoxalinol derivatives as per the method of Pailla (16). Analysis was the tumor development in these two new groups (phase II). Data À carried out by HPLC on a C18 reverse phase column with fluorescence again showed that the ( ) DOX group had fewer mice with tumors detection using an excitation E of 350 nm and an emission E of 410 nm. (Fig. 1A). The induced-expression of POX by removal of DOX in Concentration of the keto acids was calculated from linear calibration plots drinking water was shown by immunostaining for POX, which was using the internal standard method. mainly found in the cell cytoplasm consistent with its mitochon- Determination of the organic acids: lactic acid, succinic acid, and drial location (Fig. 1A). fumaric acid. Organic acids were determined by gas chromatography/mass To understand the mechanism by which POX inhibited in vivo spectrometry as per the method of Hagen (17). The characteristic fragment tumor development, we assessed proliferation and apoptosis using ions were monitored as follows; 261 m/z (lactic acid), 289 m/z (succinic the mouse tumor tissues. In TUNEL assays, consistent with in vitro acid), 287 m/z (fumaric acid), and 179 m/z (benzoic acid). Concentrations data, we found higher levels of apoptosis in (À) DOX condition of the organic acids were calculated from linear calibration plots using the internal standard method. than in (+) DOX (Fig. 1B). We also performed BrdUrd incorporation Soft agar colony formation assay. DLD-1 Tet-Off POX cells were seeded assay to assess cell proliferation. The incorporation rate was greatly at 1 Â 103 per well in six-well dishes with a bottom layer of 0.7% agar in reduced in POX cells in (À) DOX (29.4%) compared with that in (+) DMEM and a top layer of 0.35% agar in DMEM, whereas both layers DOX (65.6%) or vector cells in either condition (Fig. 1C). Because contained DOX in concentrations of 20, 1.0, 0.2, and 0.0 ng/mL, respectively. the apoptotic rate was low we suspected that cell cycle blockade/ Colonies (>0.2 mm in diameter) were counted after 10 to 14 d. At least two inhibition of proliferation was the major reason for POX’s independent assays were performed in triplicate. inhibition of tumor development and growth. Statistical analysis. Z test was used for immunohistochemical staining To characterize the effects on growth further, we performed soft data. Null hypothesis of proportion of decrease is equal to 50%. For other agar colony formation assays. When cells were kept in (+) DOX data, Fisher’s exact test and Student’s t test were used. condition (20 ng/mL), they readily formed clones. When in (À) DOX, the cloning ability of the cell was totally blocked. DOX at Results a concentration as low as 1.0 ng/mL markedly reduced clone POX is highly conserved during evolution. Using ClustalW2 formation (Fig. 1D). alignment software, we compared amino acid sequences of POX POX also caused G2 cell cycle arrest to inhibit cell growth. A protein from several species. We found homologues of POX in tumor suppressor protein exerts its inhibitory function through Homo sapiens, Mus musculus, Rattus norvegicus, Danio rerio, and either induction of apoptosis or blockade of cell cycle, or both. The Drosophila melanogaster (Supplementary Fig. S1). This finding published data from this and other laboratories clearly showed the indicates that this is highly conserved among various species role of POX in the induction of apoptosis, and the mechanism and during evolution, and suggests an important role for POX in signaling pathways involved have been suggested (11, 18–23). eukaryotic cells. However, our animal workindicated that POX might also regulate www.aacrjournals.org 6417 Cancer Res 2009;69: (16). August 15, 2009

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

Cancer Research

progression of cells in the cell cycle. Because apoptosis and not cell cycle distribution was the prominent feature of cells when POX was highly expressed in previously published studies (11, 18–20), we suspected that expression levels of POX might be critical for its roles. Thus, taking advantage of the Tet expression system, which allows the introduction of target genes dose-dependently by DOX, we determined the effects of POX at different levels on cell growth and the cell cycle. First, we confirmed that POX was indeed suppressed by DOX in a dose-dependent fashion in DLD-1 Tet-off POX cells (Fig. 2A). Then we turned on the expression of POX at different levels by varying DOX concentrations and monitored its effects on cell growth. Consistent with our previous data, cells grew normally with DOX (20 ng/mL), whereas cells died when in (À) DOX. Importantly, we found that cells neither grew nor died when cultured in medium with reduced DOX (0.2 and 0.02 ng/mL), suggesting a possible blockade in cell cycle (Fig. 2A). To investigate cell cycle distribution further, we performed flow cytometry. Based on the dose-dependent effect of DOX on POX expression, cells were treated with DOX at 100- and 1,000-fold dilutions of the usual concentration for the (+) DOX condition (0.2 and 0.02 ng/mL) for 3 days to induce POX expression at relatively low levels, with cells in (+) DOX (20 ng/mL) as control. We found that the percentages of cells in sub-G1/G0 phase were low and increased only slightly (from 2% to 4–6%); the significant changes were in S phase, which decreased from 23% to 8% to 13%, and in G2 phase, which increased from 18% to 30% to 39%, indicating a G2 cell cycle arrest by POX when it was expressed at moderate levels (Fig. 2B). Geminin is a nuclear protein that inhibits DNA replication, and has been used as a marker for G2 phase (24). Its expression was also up-regulated by POX in DLD-1 cells (Fig. 2C). To investigate further the molecular regulation of G2 phase and G2-M transition, we examined CDC2, the cyclin-dependent kinase that normally drives cells into mitosis and is the ultimate target of pathways that mediate rapid G2 arrest in response to DNA damage (25). We found that although total CDC2 did not change, the phosphorylated CDC2 at tyrosine 15 increased, whereas phosphor- ylation at theronine 161 decreased with POX expression (Fig. 2D). These changes indicated that CDC2 was in an inactive status. CDC25C, the phosphatase that removes the inhibitory phosphates from CDC2 and activates cyclinB-CDC2, was found to be down- regulated (Fig. 2D). It is likely that these changes were directly responsible for POX-induced G2 cell cycle arrest. GADDs may play a role in POX-induced G2 cell cycle arrest. The finding that POX arrested cells in G2 phase prompted us to identify pathways and genes whose expression might be respon- sible for this arrest, as well as for POX-induced apoptosis and other changes. First, we turned to cDNA microarray (Affymatrix U133A 2.0), which covers 14,500 well-characterized human genes and transcripts. We found that the expression of 814 genes had been changed >2-fold when POX is over expressed.5 Among them, the increased expression of the GADD (growth arrest and DNA damage inducible gene) family was of interest, because the most important role of this family is to cause G2 cell cycle arrest, and GADD45a, the main member of this family, is a downstream gene of p53; it directly interacts with p21 and proliferating cell nuclear antigen, both important direct regulators of the cell cycle. It also plays a role Figure 4. The reduced expression of POX in human tumor tissues. Ninety-two pairs of human cancer and normal tissues from same patient, in apoptotic induction in certain situations (26). Interestingly, most including 36 pairs from digestive tract and six pairs from kidney, were immunohistochemically stained for POX. A, the representative images were shown from tissues of colon, stomach, liver, pancreas, and kidney. Magnification, Â100. B, Z test was used to do statistical analysis (null hypothesis of proportion of decrease is equal to 50%). DT, digestive tract tissues. 5 Manuscript in preparation.

Cancer Res 2009;69: (16). August 15, 2009 6418 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

POX: A Novel Mitochondrial Tumor Suppressor genes in this family were up-regulated with POX overexpression. investigated HIF signaling. We found that HIF-1a levels were The array data revealed that GADD45a was induced by 3.2-fold, greatly reduced by POX in both normoxic and hypoxic conditions GADDh by 6.3-fold, GADDg by 3.8-fold, and GADD34 by 4.4-fold. To (Fig. 6A). As expected, the expression of its downstream gene, confirm this, we performed semiquantitive RT-PCR and Western VEGF, was also down-regulated by POX (Fig. 6A). Furthermore, we blots for these members. The POX expression of DLD-1 cells was tried to determine the mechanism by which HIF signaling was induced at different levels for 1 or 3 days. Elevation of GADD34, inhibited. As the most important oxygen sensor, the stability and GADD45a, GADDh, GADDg at the RNA level and GADD34, transcriptional activity of HIF-1a are regulated through posttrans- GADD45a, and GADD153 at the protein level was observed lational hydroxylation of specific prolyl and asparaginal residues in (Fig. 3). The detailed role of GADDs in POX’s inhibition of cell its a-subunits. These oxygen-sensitive modifications are catalyzed growth is still under investigation. But, certainly, the up-regulation by members of the 2-oxoglutarate (a-ketoglutarate) dioxygenase of GADDs may play a role in POX-induced G2 cell cycle arrest, as family (27). Recent studies have revealed that several TCA cycle well as apoptosis. intermediates and glycolytic metabolites, including succinate and The reduced expression of POX in human cancer tissues. To fumarate, inhibit PHD activity and stabilize HIF signaling (27–31). show the role/function of a gene product in cancer development We suspected that as an important linkbetween proline and progression, it is important to determine its differential metabolism and TCA cycle, a-KG, a substrate of PHD, might play expression levels in human tumors. Taking advantage of the a critical role in the down-regulation of HIF by POX (Fig. 5). availability of human tissue arrays that provide a large number and Thus, we determined the effect of POX on the cellular levels variety of cancer tissues along with normal tissue counterparts of a-KG by HPLC analysis and found increased levels of a-KG from the same patient as control, we performed immunohisto- with overexpression of POX (Fig. 6C). Furthermore, the levels of chemical staining for the expression of POX. In total, we had 92 several TCA cycle intermediates were measured by gas chroma- pairs of tumor/normal tissues from kidney, bladder, stomach, colon tography/mass spectrometry, and succinate, fumarate, and lactate and rectum, liver, pancreas, breast, prostate, ovary, brain, lung, skin, all were found to be decreased with POX expression (Fig. 6C). To etc. Strikingly, POX levels were much lower in tumor tissues when directly decide whether a-KG played a role in the inhibition of compared with normal tissues (Fig. 4). Statistical analysis showed HIF signaling by POX, we used the widely used, cell-permeating the difference significant (P < 0.05). This difference was more a-KG analogue dimethyloxalylglycine. The results showed that significant in kidney, where all six pairs showed lower POX dimethyloxalylglycine blocked the POX inhibition of HIF (Fig. 6D), expression in tumors, and digestive tract tissues, including those further supporting the important role of a-KG in the regulation from colon, rectum, stomach, liver, and pancreas (Fig. 4B). These of HIF signaling. This finding further assures the important findings indicate that POX may be related to cancer development connection between proline cycle and TCA cycle. and progression in humans, particularly in digestive tract tissues As a critical mediator of POX-mediated signaling and induction and kidney. of apoptosis, the role of reactive oxygen species (ROS)/superoxides A potential mitochondrial tumor suppressor. POX protein is in HIF inhibition with POX expression was also examined. The data located in the mitochondrial inner membrane, and has an showed that after the removal of DOX from the culture medium anaplerotic role through glutamate and a-KG for the TCA cycle and the induction of POX, HIF expression was decreased, but (Fig. 5; ref. 8). Due to this special location and function, we unlike our previously reported effects of POX on other signaling considered that POX may regulate HIF signaling, as has been pathways, the effect on HIF-1a was not reversed by MnSOD, suggested for other mitochondrial tumor suppressors (6). Thus, we suggesting ROS/superoxides were not a mediator for HIF inhibition

Figure 5. Proposed mechanisms by which POX inhibits HIF signaling. The interconversion between proline and P5C, catalyzed by POX and P5CR, respectively, forms the proline cycle. When POX expression is high, P5C, glutamate and a-KG are sequentially produced, forming an important link between proline and the TCA cycle. As an important substrate of prolyl hydroxylase, a-KG increases the hydroxylation of HIF-1a, which will be degradated through ubiquitinal and proteosomal degradation systems. This is probably the principal mechanism by which POX expression decreases HIF signaling. On the other hand, the TCA cycle is affected, as shown by decreased cellular levels of fumarate, succinate, and lactate. The decreased levels of these TCA cycle intermediates may also contribute to impaired HIF signaling. Thus, inhibition of HIF could be another mechanism, along with those previously identified, by which POX exerts its tumor-suppressing role.

www.aacrjournals.org 6419 Cancer Res 2009;69: (16). August 15, 2009

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

Cancer Research

Figure 6. The effect of POX on HIF signaling and TCA cycle intermediates. A, left, the inhibition of HIF-1a and its downstream gene VEGF was shown by Western blots in both normoxic and hypoxic conditions (1% of O2, HIF-1*, indicated longer film exposure). Right, the relative average densitometric data of four Western blots to HIF-1a in normoxic condition were also shown (B). To determine the effect of ROS/superoxide on HIF signaling, MnSOD was introduced by an adenovirus vector, which did not reverse the POX effect on the expression of HIF-1a. C, DLD-POX cells were plated with DOX. After 18 h, fresh medium with or without DOX was substituted for the plating medium. Levels of a-KG were determined by high-pressure liquid chromatography. Levels of succinate, fumerate, and lactate were determined by gas chromatography/mass spectrometry. Data are expressed as nmol/106 cells; columns, mean of at least three determinations; bars, SD (values on day 3 for each panel were statistically different from controls; P < 0.01). D, the effect of dimethyloxalylglycine on HIF-1a levels was shown by Western blots. *, statistically significant difference (P < 0.01).

(Fig. 6B). Because a-KG, a critical substrate for PHD-catalyzed patients with 22q11.2 deletion syndrome (DiGeorge hydroxylation of HIF-1a and degradation, was markedly elevated syndrome/velocardiofacial syndrome; ref. 34). Two tumor suppres- with POX overexpression, it seems likely that it is the mediator for sors have been identified in chromosome 22q11, hSNF5, and NF2 POX-dependent inhibition of HIF signaling. (35, 36). Some studies suggest the presence of another tumor suppressor gene whose alteration is responsible for the develop- ment of several types of cancer (37–39). One genetic study Discussion described ependymomas with 22q11 loci loss of heterozygosity, but The POX gene is evolutionarily conserved with a high degree of neither NF2 nor hSNF5 gene was involved (40, 41). Thus, the homology existing in drosophila, zebrafish, mouse, rat, human, etc., existence of other tumor suppressor genes at this locus was suggesting a fundamental biological role. In this present study, in suggested. It is tempting to speculate that POX may be this keeping with previously published data from this and other putative suppressor. laboratories, we established the tumor suppressor effect of POX in As a mediator of these cancer suppressive effects, we found a cancer by showing its effects on the regulation of several important novel POX-mediated mechanism in its down-regulation of HIF-1 signaling pathways: the induction of apoptosis, the blockade of the signaling. In DLD-1 POX cells, overexpression of POX increased cell cycle, and inhibition of tumor growth/development in vitro and the cellular level of a-KG, an important substrate for the in vivo. Importantly, a significant number of human tumors had hydroxylation of HIF-1a by PHDs. As a substrate for PHD, a-KG reduced expression of POX. Additionally, the data revealed that stimulates hydroxylation of HIF-1a, increases its binding to VHL POX could be a mitochondrial tumor suppressor by affecting HIF protein and causes its decrease via rapid ubiquitinylation and signaling and TCA cycle. subsequent proteasomal degradation. HIF is an antiapoptotic Tumor suppressors may lose their normal functions through factor; thus, the POX-mediated decrease in HIF signaling may various mechanisms; this loss would facilitate malignant transfor- contribute to POX-induced apoptosis and inhibition of cell and mation and cell proliferation (32). We sought genetic and tumor growth. epigenetic variations in the POX gene and its promoter using We and others previously have shown the critical role of ROS/ human tumors and normal tissues, but no definitive changes have superoxides in POX-induced apoptosis (11, 18, 19, 21). Actually, the been found thus far (occasional polymorphisms but no mutations effects of ROS on HIF could be either positive or negative, or hypermethylations). Nevertheless, the literature indicates depending on ambient oxygen levels, etc. (27). As our data showed genetic changes in POX in several human cancers. The POX gene that MnSOD had no effect on POX-mediated inhibition of HIF-1a is located at chromosome 22q11.2, a common breakpoint and a (Fig. 6D), the regulation of HIF signaling is most likely not due region subject to translocation. Deletions at this site have been directly to POX-mediated generation of superoxides. In fact, a-KG associated with a variety of human cancers (33, 34). The linkage of is the likely mediator of the decrease in HIF-1a by POX, and this POX alterations with cancer development is also supported by the decrease occurs in spite of the POX-mediated increase in ROS and observation that there is much higher frequency of malignancy in the known effect of ROS in stabilizing HIF-1a.

Cancer Res 2009;69: (16). August 15, 2009 6420 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

POX: A Novel Mitochondrial Tumor Suppressor

Thus, there are two important biochemical mechanisms related Finally, genetic or epigenetic mechanisms for the decrease or to POX-mediated stress responses. Dysregulation or loss of these loss of POX deserve further study. Although well-recognized tumor responses contributes to tumor development and/or uncontrolled suppressors such as BRCA1/2 and APC underlie familial cancer cell growth. One is redox stress resulting from increased levels of syndromes, with sporadic tumors, a substantial number of low- ROS. Another is metabolic signaling mediated by increased levels of penetrance alleles remain to be identified. Loss of these alleles in a-KG. Both may produce their effects through activation/inhibition combination may lead to malignant transformation. It is possible of well-recognized signaling pathways. Actually, these two func- that POX dysregulation, along with other carcinogenic factors, can tions may be linked and share a common denominator. For stimulate uncontrolled cell growth and eventually tumor forma- example, a-KG dehydrogenase, the enzyme converting a-KG to tion. Thus, POX as a target for cancer therapeutics, in combina- succinyl-CoA, is an important target of ROS, and is itself a tional metabolic interventions in particular, remains an attractive generator of oxidative stress (42). possibility. Mitochondrial dysfunction has long been linked to cancer, and a number of mechanisms have been suggested (3–5). The findings Disclosure of Potential Conflicts of Interest reported here suggest possible consequences in cells with POX No potential conflicts of interest were disclosed. dysregulation. POX may be up-regulated under stress conditions to retard the cell cycle and augment apoptosis on the one hand, or Acknowledgments alternatively, to provide TCA cycle intermediates and to provide Received 4/2/09; revised 6/4/09; accepted 6/6/09; published OnlineFirst 8/4/09. redox coupling of the proline cycle to alternative energy pathways Grant support: Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research. This project also has been funded in part with (43). The loss of the cancer suppressive functions of POX decreases Federal funds from the National Cancer Institute, NIH under contract no. HHSN261 POX-mediated apoptosis, removes the POX-mediated suppression 200800001. The content of this publication does not necessarily reflect the views of HIF signaling, and augments the conversion to aerobic glycolysis, or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. features of the Warburg effect. Government. National Cancer Institute-Frederickis accredited by Association for It may be pointed out that increased cancer riskin patients with Assessment and Accreditation of Laboratory Animal Care International and follows inherited deficiency of the POX has not been described. A possible the Public Health Service Policy for the Care and Use of Laboratory Animals. Animal care was provided in accordance with the procedures outlined in the ‘‘Guide for Care explanation is the redundant function of hydroxyproline oxidase in and Use of Laboratory Animals’’ (National Research Council; 1996; National Academy apoptosis (44). Furthermore, patients with this rare inborn error Press; Washington, D.C.). The costs of publication of this article were defrayed in part by the payment of page are usually identified in childhood, and careful follow-up into the charges. This article must therefore be hereby marked advertisement in accordance age range with increased cancer riskis not available. with 18 U.S.C. Section 1734 solely to indicate this fact.

References 13. Yang J, Zhu H, Murphy TL, Ouyang W, Murphy KM. gamma-dependent and -independent mechanisms. J IL-18-stimulated GADD45 h required in cytokine- Biol Chem 2006;281:2044–52. 1. Bensaad K, Vousden KH. p53: new roles in metabo- induced, but not TCR-induced, IFN-g production. 23. Hu CA, Donald SP, Yu J, et al. Overexpression lism. Trends Cell Biol 2007;17:286–91. Nature Immunology 2001;2:157–64. of proline oxidase induces proline-dependent and 2. Brahimi-Horn MC, Chiche J, Pouyssegur J. Hypoxia 14. Zhang X, Sun H, Danila DC, et al. Loss of expression mitochondria-mediated apoptosis. Mol Cell Biochem signalling controls metabolic demand. Curr Opin Cell of GADD45 g, a growth inhibitory gene, in human 2007;295:85–92. Biol 2007;19:223–9. pituitary adenomas: implications for tumorigenesis. 24. Zhu W, Chen Y, Dutta A. Rereplication by depletion 3. Hagland H, Nikolaisen J, Hodneland LI, Gjertsen BT, J Clin Endocrinol Metab 2002;87:1262–7. of geminin is seen regardless of p53 status and Bruserud O, Tronstad KJ. Targeting mitochondria in the 15. Zou C, Guan Y, Zou C, et al. N-(4-hydroxyphenyl) activates a G2/M checkpoint. Mol Cell Biol 2004;24: treatment of human cancer: a coordinated attack retinamide (4-HPR) modulates GADD45 expression in 7140–50. against cancer cell energy metabolism and signalling. radiosensitive bladder cancer cell lines. Cancer Lett 25. StarkGR, Taylor WR. Control of the G2/M transition. Exp Opin Ther Targets 2007;11:1055–69. 2002;180:131–7. Mol Biotechnol 2006;32:227–48. 4. Ristow M. Oxidative metabolism in cancer growth. 16. Pailla K, Blonde-Cynober F, Aussel C, De Bandt JP, 26. Liebermann DA, Hoffman B. Gadd45 in stress Curr Opin Clin Nut Metab Care 2006;9:339–45. Cynober L. Branched-chain keto-acids and pyruvate in signaling. J Mol Signal 2008;3:15. 5. Gottlieb E, Tomlinson IP. Mitochondrial tumour blood: measurement by HPLC with fluorimetric detec- 27. Verma A. Oxygen-sensing in tumors. Curr Opin Clin suppressors: a genetic and biochemical update. Nat tion and changes in older subjects. Clin Chem 2000;46: Nut Metab Care 2006;9:366–78. Rev Cancer 2005;5:857–66. 848–53. 28. Bruegge K, Jelkmann W, Metzen E. Hydroxylation of 6. Kowaloff EM, Phang JM, Granger AS, Downing SJ. 17. Hagen T, Korson MS, Sakamoto M, Evans JE. A GC/ hypoxia-inducible transcription factors and chemical Regulation of proline oxidase activity by lactate. Proc MS/MS screening method for multiple organic acid- compounds targeting the HIF-a hydroxylases. Curr Med Natl Acad Sci U S A 1977;74:5368–71. emias from urine specimens. Inter J Clin Chem 1999;283: Chem 2007;14:1853–62. 7. Phang JM. The regulatory functions of proline and 77–88. 29. Hewitson KS, Lienard BM, McDonough MA, et al. pyrroline-5-carboxylic acid. Curr Top Cell Reg 1985;25: 18. Liu Y, Borchert GL, Donald SP, et al. MnSOD inhibits Structural and mechanistic studies on the inhibition of 91–132. proline oxidase-induced apoptosis in colorectal cancer the hypoxia-inducible transcription factor hydroxylases 8. Phang JM, Pandhare J, Liu Y. The metabolism of cells. Carcinogenesis 2005;26:1335–42. by tricarboxylic acid cycle intermediates. J Biol Chem proline as microenvironmental stress substrate. J Nut 19. Liu Y, Borchert GL, Surazynski A, Hu CA, Phang JM. 2007;282:3293–301. 2008;138:2008–15S. Proline oxidase activates both intrinsic and extrinsic 30. Koivunen P, Hirsila M, Remes AM, Hassinen IE, 9. PolyakK, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. A pathways for apoptosis: the role of ROS/superoxides, Kivirikko KI, Myllyharju J. Inhibition of hypoxia- model for p53-induced apoptosis. Nature 1997;389:300–5. NFAT and MEK/ERK signaling. Oncogene 2006;25: inducible factor (HIF) hydroxylases by citric acid 10. Donald SP, Sun XY, Hu CA, et al. Proline oxidase 5640–7. cycle intermediates: possible links between cell meta- encoded by p53-induced gene-6 catalyzes the generation 20. Maxwell SA, Rivera A. Proline oxidase induces bolism and stabilization of HIF. J Biol Chem 2007;282: of proline-dependent reactive oxygen species. Cancer apoptosis in tumor cells, and its expression is frequently 4524–32. Res 2001;61:1810–5. absent or reduced in renal carcinomas. J Biol Chem 31. Lu H, Dalgard CL, Mohyeldin A, McFate T, Tait AS, 11. Liu Y, Borchert GL, Surazynski A, Phang JM. Proline 2003;278:9784–9. Verma A. Reversible inactivation of HIF-1 prolyl oxidase, a p53-induced gene, targets COX-2/PGE2 21. Rivera A, Maxwell SA. The p53-induced gene-6 (proline hydroxylases allows cell metabolism to control basal signaling to induce apoptosis and inhibit tumor growth oxidase) mediates apoptosis through a calcineurin- HIF-1. J Biol Chem 2005;280:41928–39. in colorectal cancers. Oncogene 2008;27:6729–37. dependent pathway. J Biol Chem 2005;280:29346–54. 32. Payne SR, Kemp CJ. Tumor suppressor genetics. 12. Fishel ML, RabikCA, Bleibel WK, Li X, Moschel RC, 22. Pandhare J, Cooper SK, Phang JM. Proline oxidase, a Carcinogenesis 2005;26:2031–45. Dolan ME. Role of GADD34 in modulation of cisplatin proapoptotic gene, is induced by troglitazone: evidence 33. Handel-Fernandez ME, Nassiri M, Arana M, et al. cytotoxicity. Bioch Pharmacol 2006;71:239–47. for both peroxisomal proliferator-activated receptor Mapping of genetic deletions on the long arm of www.aacrjournals.org 6421 Cancer Res 2009;69: (16). August 15, 2009

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

Cancer Research

in human pancreatic adenocarcinomas. evidence of INI1hSNF5 (SMARCB1) and PARVG point Spectrum of hSNF5/INI1 somatic mutations in human Anticancer Res 2000;20:4451–6. mutations in oligodendroglial neoplasms. Cancer Genet cancer and genotype-phenotype correlations. Hum Mol 34. McDonald-McGinn DM, Reilly A, Wallgren-Pettersson Cytogenet 2005;160:169–73. Genet 1999;8:2359–68. C, et al. Malignancy in chromosome 22q11.2 deletion 38. KruzelockRP, Cuevas BD, Wiener JR, et al. Functional 42. Tretter L, Adam-Vizi V. a-ketoglutarate dehydroge- syndrome (DiGeorge syndrome/velocardiofacial syn- evidence for an ovarian cancer tumor suppressor gene nase: a target and generator of oxidative stress. drome). Am J Med Genet 2006;140:906–9. on chromosome 22 by microcell-mediated chromosome Philosoph Trans Royal Soc London 2005;360:2335–45. 35. Schmucker B, Tang Y, Kressel M. Novel alternatively transfer. Oncogene 2000;19:6277–85. 43. Hagedorn CH, Phang JM. Catalytic transfer of hydride spliced isoforms of the neurofibromatosis type 2 tumor 39. Manda R, Kohno T, Hamada K, Takenoshita S, ions from NADPH to oxygen by the interconversions of suppressor are targeted to the nucleus and cytoplasmic Kuwano H, Yokota J. Absence of hSNF5/INI1 mutation proline and y 1-pyrroline-5-carboxylate. Arch Biochem granules. Hum Mol Genet 1999;8:1561–70. in human lung cancer. Cancer Lett 2000;153:57–61. Biophy 1986;248:166–74. 36. Versteege I, Sevenet N, Lange J, et al. Truncating 40. Huang B, StarostikP, Kuhl J, Tonn JC, Roggendorf W. 44. Cooper SK, Pandhare J, Donald SP, Phang JM. A novel mutations of hSNF5/INI1 in aggressive paediatric Loss of heterozygosity on chromosome 22 in human function for hydroxyproline oxidase in apoptosis cancer. Nature 1998;394:203–6. ependymomas. Acta Neuropathol 2002;103:415–20. through generation of reactive oxygen species. J Biol 37. Alonso ME, Bello MJ, de Campos JM, et al. No 41. Sevenet N, Lellouch-Tubiana A, Schofield D, et al. Chem 2008;283:10485–92.

Cancer Res 2009;69: (16). August 15, 2009 6422 www.aacrjournals.org

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Published OnlineFirst August 4, 2009; DOI: 10.1158/0008-5472.CAN-09-1223

Proline Oxidase Functions as a Mitochondrial Tumor Suppressor in Human Cancers

Yongmin Liu, Gregory L. Borchert, Steven P. Donald, et al.

Cancer Res 2009;69:6414-6422. Published OnlineFirst August 4, 2009.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-09-1223

Cited articles This article cites 44 articles, 11 of which you can access for free at: http://cancerres.aacrjournals.org/content/69/16/6414.full#ref-list-1

Citing articles This article has been cited by 7 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/69/16/6414.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/69/16/6414. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research.