View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Elsevier - Publisher Connector

SUMO-Specific 1 Is Essential for Stabilization of HIF1a during Hypoxia

Jinke Cheng,1,3 Xunlei Kang,2,3 Sui Zhang,1 and Edward T.H. Yeh1,2,* 1Department of Cardiology, the University of Texas M.D. Anderson Cancer Center 2Center for Cardiovascular Research, Brown Foundation Institute of Molecular Medicine The University of Texas, Houston Health Science Center, Houston, TX 77030, USA 3Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, P. R. China *Correspondence: [email protected] DOI 10.1016/j.cell.2007.08.045

SUMMARY these can alter their cellular localization and bio- logical activity. SUMOylation is a dynamic process, catalyzed SUMO conjugation is a dynamic process, in that it can by SUMO-specific ligases and reversed by Sen- be readily reversed by a family of Sentrin/SUMO-specific trin/SUMO-specific (SENPs). The proteases (SENPs) (Hay, 2005; Yeh et al., 2000). Six physiologic consequences of SUMOylation SENPs have been identified in human, each with different and deSUMOylation are not fully understood. cellular location and substrate specificity (Yeh et al., Here we investigate the phenotypes of mice 2000). They can be divided into three subfamilies on the basis of their , cellular location, and lacking SENP1 and find that SENP1/ embryos substrate specificity. The first subfamily consists of show severe fetal anemia stemming from defi- SENP1 and SENP2, which have broad substrate specific- cient (Epo) production and die ity. The second subfamily consists of SENP3 and SENP5, midgestation. We determine that SENP1 con- both of which are nucleolar proteins with preferences for trols Epo production by regulating the stability SUMO-2/3 (Di Bacco et al., 2006; Gong and Yeh, 2006). of hypoxia-inducible factor 1a (HIF1a) during The third subfamily consists of SENP6 and SENP7, which hypoxia. Hypoxia induces SUMOylation of HIF1a, have an extra loop in their catalytic domains. Although which promotes its binding to a ubiquitin SENPs are known to reverse SUMOylation in many ligase, von Hippel-Lindau (VHL) , through different systems, their physiological role has not been a -independent mecha- precisely defined. nism, leading to its ubiquitination and degra- SENP1, a nuclear SUMO protease, has been shown to regulate androgen transactivation by targeting dation. In SENP1/ MEFs, hypoxia-induced 1 and to induce c-Jun activity through of HIF1a-dependent such deSUMOylation of p300 (Cheng et al., 2004, 2005). Yama- as vascular endothelial growth factor (VEGF) guchi et al. studied mice derived from an ES cell line with and glucose transporter 1 (Glut-1) is markedly a retroviral vector that had been randomly inserted into the reduced. These results show that SENP1 plays enhancer region on the SENP1 . This random inser- a key role in the regulation of the hypoxic tion reduced expression of the SENP1 transcript, causing response through regulation of HIF1a stability the mice to die between E12.5 and E14.5 (Yamaguchi and that SUMOylation can serve as a direct et al., 2005). Although no specific histological abnormali- signal for ubiquitin-dependent degradation. ties were found in the E13.5 embryos, there was a hint of an abnormality in the development of blood vessels in the placenta. Thus, it remains unclear how SENP1 contrib- INTRODUCTION utes to normal development. Therefore, we generated SENP1 knockout mice to de- Conjugation of small ubiquitin-related modifier protein lineate the contribution of SENP1 in development. Inacti- (SUMO) to a large number of substrates has been sug- vation of the SENP1 gene causes severe fetal anemia in gested to regulate numerous cellular processes from midgestation as a result of deficient Epo production. yeast to mammal (Hay, 2005; Yeh et al., 2000). Most Epo is essential for growth and survival of erythroid pro- SUMO targets are in the nucleus; they include transcrip- genitors during differentiation into red cells (Wu et al., tion factors, transcriptional coregulators, and chromo- 1995). We found that SENP1 controls Epo production by some-remodeling regulators (Gill, 2004). SUMOylation of regulating the stability of hypoxia-inducible factor 1a

584 Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. (HIF1a). Hypoxia induces HIF1a SUMOylation, which mia was observed after E13.5 in SENP1/ embryos, we promotes HIF1a degradation through a VHL- and pro- reasoned that these embryos likely had a defect in defin- teasome-dependent mechanism. SENP1 deconjugates itive erythropoiesis. Indeed, the number of nucleated SUMOylated HIF1a and allows HIF1a to escape cells in the fetal livers of SENP1/ embryos was markedly degradation during hypoxia. These results reveal an decreased compared to their wild-type littermates important physiological role of SENP1 in the hypoxic (Figure 1D). Histological examination of fetal liver sections response through regulation of HIF1a stability and that also revealed a marked decrease in the number of eryth- SUMOylation can also target a protein for ubiquitination ropoietic foci in SENP1/ embryos compared with their and degradation. wild-type littermates (Figure 1E).

RESULTS Increased Apoptosis in Erythroid Precursors of SENP1/ Embryos Generation of SENP1 Knockout Mice We next examined hematopoiesis in the fetal livers of both To generate SENP1 knockout mice, a gene-trapped vec- wild-type and SENP1/ embryos at E13.5 by flow cytom- tor was inserted into the mouse SENP1 open reading etry using markers, including CD34, CD44, c-kit, and Ter- frame at codon 310 (Figure S1A). Specifically, the inserted 119, characteristic of different types of hematopoietic b-geo (b-galactosidase/neomycin-resistance fusion pro- cells and developmental stages (Kondo et al., 2003; tein) was fused into the N terminus of SENP1 at codon Neubauer et al., 1998). There was no significant difference 310 to generate SENP1 (1-309)-b-geo fusion protein, between SENP1/ embryos and wild-type littermates, which lacks the C-terminal catalytic domain of SENP1 except that the population of Ter-119+, which denotes (Figure S1A). This disruption was confirmed by the ab- committed erythropoietic precursors, was markedly re- sence of transcripts that encode the catalytic domain of duced in SENP1/ fetal livers (Figure S2A). These data SENP1 in SENP1/ embryos (Figure S1B). The overall suggest that erythropoiesis was abnormal in SENP1/ SUMOylation pattern in lysates of embryos revealed an in- embryos as a result of a defect in erythroid differentiation. crease in high-molecular-weight SUMO-1 and SUMO-2/3 To determine at which stage of erythroid differentiation conjugates in SENP1/ embryos in comparison with SENP1 plays a crucial role, we compared the abilities of wild-type or SENP1+/ embryos (Figure S1C). In addition, cells derived from wild-type and SENP1/ E13.5 fetal processing of the C terminus of SUMO-1 but not SUMO-2/ livers to form erythroid colony-forming units (CFU-e) and 3 appeared to be decreased in SENP1/ embryos more immature erythroid burst-forming units (BFU-e) (Figure S1C). These results indicate that disruption of the (Wu et al., 1995). Although fetal livers from SENP1/ em- SENP1 locus reduced deSUMOylation in SENP1/ em- bryos contained erythroid progenitors, the relative number bryos. of CFU-e arising from SENP1/ fetal liver was over 40% smaller than that from wild-type fetal liver (Figure 2A). Severe Anemia in SENP1/ Embryos However, there was no significant difference in the num- Heterozygous mice carrying the inactivated SENP1 gene bers of the BFU-e between SENP1/ and wild-type fetal appeared normal and fertile. However, no live SENP1/ livers (Figure 2A). Additionally, we observed no significant mice were found among offsprings of SENP1+/ inter- difference in the number of colony-forming unit granulo- crosses, indicating that the SENP1/ mutation leads to cyte-macrophage (CFU-GM) and pluripotent hematopoi- embryonic lethality. Examination of SENP1/ embryos etic stem cells (CFU-GEMM) between SENP1/ and at different stages of development revealed that the major- wild-type fetal livers (Figure S2B). These results indicate ity died between days 13 and 15 of gestation (Figure 1A). that a developmental defect in CFU-e is a prominent fea- The SENP1/ embryos appeared paler and smaller than ture in the SENP1/ embryo. their wild-type or SENP1+/ littermates (Figure 1B). The The reduced ratio of CFU-e to BFU-e progenitors and most dramatic morphological abnormality of these the decrease in the number of Ter-119+ erythroid cell sug- SENP1/ embryos was severe fetal anemia. In particular, gested a reduction in the net growth of SENP1/ erythroid SENP1/ embryos had more than 75% fewer erythro- progenitors resulting from either decreased cell prolifera- cytes than their wild-type or SENP1+/ littermates at tion or increased apoptosis. No differences were observed E15.5 (Figure 1C). in the proliferation of fetal liver cells between SENP1/ embryos and their wild-type littermates, as shown by Ki- Defective Definitive Erythropoiesis in SENP1/ 67 staining (upper panel, Figure S2C). However, TUNEL Embryos staining showed that the number of TUNEL-positive cells During normal murine development, erythropoiesis is initi- was much greater in the liver sections from SENP1/ em- ated in the yolk sac at around E8 (primitive erythropoiesis), bryos than in those of their wild-type littermates (lower and then it is initiated in the aorta-gonadmesonephros panel, Figure S2C). To validate further, we stained fetal (AGM) region. At midgestation, fetal liver becomes the ma- liver cells with the anti-Ter-119 antibody and then analyzed jor hematopoietic organ with hematopoietic activity start- cells by flow cytometry. This showed that the percentage ing around E11.0 to E12.5 until 1 week postnatal (definitive of Ter-119-positive cells was decreased in SENP/ em- erythropoiesis) (Godin and Cumano, 2002). Because ane- bryos, from 81% in E13.5 wild-type fetal livers to 37% in

Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. 585 Figure 1. Severe Anemia in SENP1/ Embryos (A) The ratios of the observed live and dead (in parentheses) SENP1/ embryos to the total number of embryos analyzed at different stages of embryonic development. (B) Appearance of SENP1+/+ and SENP1/ embryos at E15.5. The SENP1/ embryo and yolk sac were paler and smaller than those of the wild-type embryo and appeared to con- tain fewer red blood cells in major blood ves- sels. (C) Relative numbers of red blood cells in pe- ripheral blood in E15.5 wild-type (+/+, n = 5), heterozygote (+/, n = 9), and SENP1 (/, n = 4) embryos. Results shown are means ± standard deviation (SD). A significant decrease in the number of red blood cells was found in SENP1 (/) embryos (p < 0.007) when com- pared with those in wild-type or heterozygote embryos. (D) Total number of nucleated cells per fetal liver in E13.5 (+/+, n = 4; /, n = 3), E14.5 (+/+, n = 4; /, n = 4), and E15.5 embryos (+/+, n = 5; /, n = 3). Results shown are means ± SD. Significant differences between wild-type and mutant embryos were found at E14.5 (p < 0.015) and E15.5 (p < 0.004). (E) Hematoxylin- and Eosin-stained sections of fetal liver from E12.5 SENP1+/+ and / em- bryos. The SENP1/ fetal liver showed a marked decrease in the number of erythro- poietic foci and increase in apoptotic cells.

their SENP1/ littermates (data not shown). Among the age in the Epo signaling pathway. To assess these possi- Ter-119-positive cells, about 34.5% of SENP1/ cells bilities, we first examined the expression of genes in- were stained by TUNEL; in contrast, few TUNEL-positive volved in the Epo signaling pathway (specifically, EpoR, cells (1.9%) were observed from the wild-type liver Jak2, and STAT5) and other growth factors associated (Figure 2B). These results suggested that the erythroid with erythroid progenitors in fetal livers. A dramatic reduc- cells in fetal liver were more apoptotic in SENP1/ em- tion in the level of Epo mRNA expression was shown in bryos than in their wild-type counterparts, which accounts SENP1/ fetal livers (Figure 2C). In contrast, the expres- for the decrease in CFU-e progenitors, smaller fetal liver sion of EpoR, Jak2, STAT5, and growth factors c-kit and size, and anemia in SENP1/ embryos. SCF was not significantly different between SENP1/ and wild-type fetal livers (Figure 2C). The reduced Epo expression in E12.5 fetal liver cells of SENP1/ embryos Epo Production Is Reduced in SENP1/ was further confirmed by immunohistochemistry (Fig- Fetal Liver ure 2D). These results demonstrated that Epo production The hematopoietic defect in SENP1/ embryos de- was significantly reduced in SENP1/ fetal livers. scribed above was similar to that observed in Epo/ em- bryos or in embryos with mutation of the genes critical to the Epo signaling pathway (Neubauer et al., 1998; Parga- Epo Prevents Apoptosis of SENP1/ nas et al., 1998; Socolovsky et al., 1999; Wu et al., 1995). Fetal Liver Cells Since fetal liver erythroid progenitors depend on Epo for To determine whether apoptosis of cells in the fetal livers growth and survival during terminal differentiation into of SENP1/ embryos was caused by a deficiency in Epo red blood cells (Wu et al., 1995), we reasoned that the in- production, we examined the ability of Epo to prevent creased apoptosis of erythroid cells in SENP1/ fetal liver apoptosis of the erythroid progenitors from SENP1/ could be due to either a defect in Epo production or block- fetal liver. As shown by rates of apoptosis in vitro, E13.5

586 Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. Figure 2. SENP1/ Erythroid Progeni- tors Undergo Apoptosis due to Epo Defi- ciency (A) Analysis of CFU-e- and BFU-e-forming abil- ity of fetal liver cells from E13.5 SENP1+/+ and SENP1/ embryos. Results shown are means ± SD determined from three embryos. The number of CFU-e from liver cells of SENP1/ embryos was significantly less than that from SENP1+/+ embryos (p < 0.05). (B) Liver cells isolated from E13.5 SENP1+/+ and SENP1/ embryos were stained with Ter-119 antibody and analyzed by the TUNEL procedure. Histograms show TUNEL staining profile of the Ter-119-positive population iden- tified by flow cytometry. The percentages in the histograms are the percentages of TU- NEL-positive cells. (C) RT–PCR analysis of a variety of genes that are involved in erythroid differentiation in fetal livers of SENP1+/+ and SENP1/ embryos at E11.5. Samples were amplified for 36, 39, and 42 cycles for Epo; 25, 28, and 32 cycles for EpoR, c-kit, SCF, Jak2, and STAT5. b-Actin mRNA levels (amplified for 18, 21, and 24 cy- cles) were measured as control. (D) Fetal liver sections from E12.5 SENP1+/+ and SENP1/ embryos were stained with anti-Epo antibody (brown). (E and F) Fetal liver cells isolated from E13.5 SENP1+/+ and SENP1/ embryos were cul- tured for 24 hr in the absence or presence of Epo (5 U/ml). Cell numbers were counted un- der microscope (E). Results shown in (E) are means ± SD determined from three embryos. TUNEL-positive cells were quantitated by flow cytometry. Percentages in histograms are the percentages of TUNEL-positive cells (F). wild-type fetal liver cells, which were grown in a medium ment with hypoxia (Figure 3A), indicating that SENP1 lacking Epo, showed a reduction in cell number and in- played an important role in Epo production under the hyp- crease in apoptosis similar to those observed in fetal liver oxia condition. cells of their SENP1/ littermates. Addition of Epo to the Since SENP1 regulates the transcription of numerous culture medium prevented reduction in cell number and genes by targeting transcription factors or coregulators apoptosis in SENP1/ fetal liver cells similar to their (Cheng et al., 2004, 2005), we next examined whether wild-type littermates (Figures 2E and 2F). These results Epo transcription was also regulated by SENP1. In this demonstrated that the increased apoptosis of erythroid study, the luciferase reporter gene driven by the Epo pro- progenitors was due to deficiency in Epo production in moter plus enhancer (Epo-Luc) was transfected into Hep SENP1/ fetal liver. 3B cells with SENP1 or SENP1 catalytic inactive mutant (SENP1m), in which a conserved amino acid, SENP1 Regulates Epo Transcription through 603, in the catalytic domain of SENP1 was substituted HIF1a with alanine. In the presence of hypoxia, SENP1, but not The Epo gene is expressed primarily in fetal liver and adult the SENP1 mutant, significantly induced Epo reporter and is regulated in response to oxygen availability gene transcription (Figure 3B), suggesting that SENP1 (Ebert and Bunn, 1999). Since SENP1 is expressed in regulated Epo transcription and the regulation required mouse fetal liver at midgestation stage (Figure S3), we rea- the deSUMOylation activity of SENP1. Interestingly, with- soned that SENP1 might directly regulate Epo expression out hypoxia treatment, SENP1 had only marginal effect on in fetal liver. A hepatoma cell line, Hep 3B, was used to de- Epo transcription. termine whether SENP1 regulated Epo production. Two Since HIF1a is a major for hypoxia- SENP1-specific siRNAs were able to efficiently knock regulated Epo expression (Giaccia et al., 2004), we asked down SENP1 expression (Figure S4) and significantly re- whether HIF1a was required for SENP1-regulated Epo duced Epo expression in Hep 3B cells in response to treat- transcription. Coexpression of SENP1 markedly enhanced

Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. 587 Figure 3. SENP1 Regulates Epo Produc- tion through HIF1a (A) ELISA analysis of Epo production in Hep 3B cells. Data are presented as means ± SD of the results of three independent experiments for Figures 3A, 3B, 3C, and 3D. (NS = nonspecific siRNA). (B) SENP1 enhanced Epo transcription in- duced by hypoxia. The indicated reporter gene, HIF1a, and SENP1 expression plasmids were cotransfected into Hep 3B cells. The cells

were treated with hypoxia (1% O2) for 12 hr be- fore luciferase assay. The Epo reporter gene with mutation of HIF1a-binding sites on the Epo enhancer is indicated by mEpo-Luc. SENP1m is the catalytically inactive SENP1. (C) SENP1 enhanced HIF1a-dependent Epo transcription. The indicated reporter gene, HIF1a, and SENP1 expression plasmids were cotransfected into Hep 3B cells. (D) SENP1 was essential for HIF1a-dependent Epo transcription. Epo-Luc and the indicated siRNA expression plasmids were cotrans- fected into Hep 3B cells without or with HIF1a plasmids.

HIF1a-dependent Epo transcription (Figure 3C). However, Furthermore, the half-life of HIF1a protein was markedly mutation of HIF1a-binding sites on the Epo enhancer reduced in the SENP1/ MEF cells under the hypoxia (mEpo-Luc) completely abolished SENP1 activity (Fig- condition in a pulse-chase experiment (Figure 4C). Nor- ure 3C). Furthermore, silencing of endogenous SENP1 moxia-induced HIF1a degradation is dependent on its in- expression in Hep 3B cells using SENP1 siRNA (SENP1- ternal oxygen-depend-degradation (ODD) domain (Huang si1 and SENP1-si2) also reduced Epo transcription in et al., 1998). We then determined whether the ODD do- response to HIF1a by 80% (Figure 3D). These results main was a direct target of SENP1 regulation of HIF1a sta- strongly suggested that Epo expression was regulated bility under hypoxia condition. We used the HIF1a ODD by SENP1 through its regulation of HIF1a activity. domain (344-698) fused Gal4 (pG4-ODD-VP16), which was previously demonstrated to have normoxic- and SENP1 Is Essential for Ensuring HIF1a Stability VHL-dependent degradation and activity (Huang et al., during Hypoxia 1998; Maxwell et al., 1999). Indeed, coexpression of It is well-known that regulation of HIF1a occurs mostly at SENP1 markedly enhanced hypoxia-induced ODD activ- the protein level. In SENP1/ mice, HIF1a protein level ity. However, hypoxia-induced activity of the ODD domain was significantly decreased in a section of the SENP1/ was completely abolished by SENP1-siRNA (Figure 4D). fetal liver compared with the SENP1+/+ littermate These results strongly indicated that SENP1 was essential (Figure 4A). However, HIF1a mRNA levels in fetal livers for HIF1a stabilization under hypoxia condition. of SENP1/ or wild-type embryos were similar, suggest- Consistent with the decrease in HIF1a expression dur- ing that the decrease in HIF1a protein level resulted from ing hypoxia, HIF1a activity was also significantly reduced protein degradation but not a reduction of HIF1a tran- in SENP1/ MEF cells as measured by the response of scription in SENP1/ embryos (Figure S5). the HRE-luciferase reporter gene to hypoxia in SENP1/ HIF1a is degraded by a proteasome-dependent mech- and wild-type MEF cells (Figure 4E). As HIF1a is a major anism under normoxia (Huang et al., 1998; Maxwell et al., transcription factor in the regulation of a large number of 1999; Wang et al., 1995). Hypoxia was believed to stabilize hypoxia-inducible genes, we also showed that hypoxia-in- HIF1a protein and therefore increased its activity (Giaccia duced transcription of VEGF and Glut-1 genes was dra- et al., 2004; Huang et al., 1998; Maxwell et al., 1999). We matically decreased in SENP1/ MEF cells (Figure 4F). hypothesized that regulation of HIF1a protein to hypoxia This defect was similar to the response of HIF1a/ ES was defective in SENP1/ cells. To test this possibility, upon hypoxia exposure (Iyer et al., 1998; Ryan et al., we determined changes in HIF1a protein level in 1998). We further confirmed that this defect in SENP1/ SENP1/ and wild-type MEF cells when exposed to hyp- MEF cells was mediated by HIF1a, as transfected HI- oxia. As shown in Figure 4B, hypoxia markedly increased F1aSM (SUMOylation mutant) could restore hypoxia-in- the protein level of HIF1a in the wild-type MEF cells, but duced expression of the VEGF gene in the transfected the increase was much less in the SENP1/ MEF cells. cells (Figure 4G). These results indicated that SENP1

588 Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. Figure 4. Defect in Hypoxia-Induced Sta- bilization of HIF1a in SENP1/ Embryos (A) HIF1a expression was decreased in SENP1/ fetal liver. Fetal liver sections from E12.5 SENP1+/+ and SENP1/ embryos were stained with anti-HIF1a antibody (brown). (B) Hypoxia-induced HIF1a protein expression was decreased in SENP1/ MEF cells. SENP1+/+ and / MEF cells were treated

with hypoxia (1% O2) for 3 hr before harvest. The whole-cell lysate (WCL) was analyzed by western blotting with anti-HIF1a and actin anti- bodies. (C) The half-life of HIF1a protein under hypoxia was decreased in SENP1/ MEF cells. (D) SENP1 was essential for hypoxia-induced HIF1a ODD(344–698) activity. 293 cells were transfected with pGal4-VP16 or pGal4- ODD(344–698)-VP16 plus other plasmids as in- dicated. Cells were incubated for 12 hr in nor-

moxia or hypoxia (1% O2) before harvest. The data are presented as the corrected (by internal control) pGal4-ODD-VP16 luciferase activity that normalized to the counts of pGal4-VP16. (E) Hypoxia-induced HIF1a activity was signifi- cantly reduced in SENP1/ MEF cells. Data are presented as means ± SD of three indepen- dent experiments. (F) Hypoxia-induced expression of VEGF and Glut-1 was reduced in SENP1/ MEF cells. Expression of VEGF, Glut-1, and Actin-b was determined in SENP1+/+ or / MEF cells by RT-PCR. (G) HIF1a restored hypoxia-induced expres- sion of VEGF in SENP1/ MEF cells. SENP1/ MEF cells were transfected with HIF1a SM (SUMOylation mutant) or vector con-

trol plasmids and treated with hypoxia (1% O2) for different times as indicated. Expression of VEGF and Actin-b was determined by RT-PCR. played a critical role in regulating the stability and activity The simultaneous accumulation of SUMOylated HIF1a of HIF1a protein during hypoxia. and the decrease in HIF1a stability in SENP1/ cells dur- ing hypoxia prompted us to study whether SUMOylation might lead to HIF1a degradation. Indeed, SUMOylated SUMOylated HIF1a Is Degraded in a Proteasome- bands of HIF1a resulted from coexpression of HIF1a Dependent Manner and HA-SUMO-1 and were difficult to detect in the ab- Since SENP1 could deSUMOylate HIF1a, SENP1 might sence of a proteasome inhibitor MG132 but readily detect- be directly responsible for the decrease in HIF1a activity able when both SENP1 siRNA and MG132 were used and stability in the SENP1/ embryo (Figures 5A and (Figure 5D). These results were further confirmed by ex- S6). The effect of deSUMOylation on HIF1a activity was amining endogenous HIF1a in SENP1/ and wild-type directly confirmed by using HIF1a SUMOylated site mu- MEF cells with exposure to both hypoxia and MG132. tants: K391R, K477R, and K391/477R (SM) (Bae et al., As expected, hypoxia-induced SUMOylated HIF1a was 2004). As shown in Figure 5B, mutation of the two readily detected in SENP1/ MEF but not in wild-type SUMOylation sites of HIF1a (SM) significantly increased MEF cells. Furthermore, MG132 treatment markedly in- the transcriptional activity of HIF1a and also reduced the creased hypoxia-induced SUMOylated bands of HIF1a ability of SENP1 to enhance HIF1a-dependent Epo tran- in SENP1/ MEF cells, suggesting that the level of scription. Furthermore, hypoxia-induced accumulation of SUMOylated HIF1a was controlled by a proteasome-de- SUMOylated HIF1a only occurred in SENP1/ MEF cells pendent mechanism (Figure 5E, upper panel). Meanwhile, but not in wild-type cells (Figure 5C). Interestingly, this ac- HIF1a protein level was significantly increased in SENP1 cumulation did not appear in SENP2/ MEF cells wild-type MEF cells but not in the SENP1/ MEF cells (Figure S7), suggesting that SENP1 specifically targeted with exposure to hypoxia (Figure 5E, lower panel). Taken HIF1a for deSUMOylation (Figure 5C). together, these results suggested that SUMOylated

Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. 589 Figure 5. SUMOylated HIF1a Accu- mulates in SENP1/ MEF Cells and Undergoes Proteasomal-Dependent Degradation under Hypoxia (A) SENP1 deconjugated SUMOylated HIF1a in vitro. SUMOylated GST-ODD(344–698) PM re- combinant protein was produced by in vitro SUMOylation. Flag-SENP1 or SENP1m was generated by in vitro translation. SUMOylated GST-ODDPM and SENP1 (or SENP1m) were incubated for 1 hr at 37C. The reaction mix- tures were detected by western blot with anti- HIF1a (left panel), anti-SUMO-1 (right panel), and anti-Flag antibodies (bottom panel). (B) Mutation of SUMOylation sites increased HIF1a-dependent Epo transcription and re- duced HIF1a response to SENP1. Epo-Luc and the indicated plasmids were cotransfected into Hep 3B cells. Data are presented as means ± SD of three independent experiments. (C) SUMOylated HIF1a accumulated in SENP1/ MEF cells but not in wild-type or SENP2/ MEF cells after exposure to hypoxia. Wild-type (+/+), SENP1/,orSENP2/ MEF cells were treated with or without hypoxia (1%

O2) for 4 hr as indicated. HIF1a was immuno- precipitated with anti-HIF1a antibody from these cell lysates. The precipitates were immu- noblotted (IB) with anti-SUMO-1 antibody (top panel). Asterisk indicates IgG band. (D) SUMOylated HIF1a level was controlled by SENP1 and proteasome-dependent degrada- tion. COS-7 cells were transfected with indi- cated plasmids and then treated without or with MG132 (10 mM) for 4 hr before harvesting and immunoprecipitated with anti-HIF1a (IP). Bound proteins were detected by anti-HA and anti-HIF1a immunoblotting (IB). WCLs were im- munoblotted (IB) with anti-HA. Asterisk indi- cates IgG band. (E) SUMOylated HIF1a accumulated in SENP1/ MEF cells after exposure to hypoxia and underwent proteasome-dependent degradation. SENP1+/+ / or MEF cells were treated by hypoxia (1% O2) and MG132 (10 mM) for 4 hr as indicated. HIF1a was immunoprecipitated with anti-HIF1a antibody from cell lysates. The precipitates were immunoblotted (IB) with anti-SUMO-1 and -HIF1a antibodies. Asterisk indicates IgG band.

HIF1a was degraded in a proteasome-dependent manner 1999). SUMOylated HIF1a was easily detected in VHL during hypoxia in the absence of SENP1. null RCC4 cells even without MG132 treatment (lane 2 ver- sus 3 in top panel, Figure 6B). In contrast, this band was only weakly detectable in RCC4/VHL cells exposed to Degradation of SUMOylated HIF1a Is Dependent hypoxia and MG132 treatment. Consistent with other re- on VHL ports, hypoxia-induced increase in HIF1a protein was The above results suggest that SUMO and SENP1 might only observed in VHL-restored RCC4 cells (bottom regulate ubiquitination of HIF1a. As shown in Figure 6A, panel, Figure 6B). These results suggested that SUMOy- ubiquitination of HIF1a was significantly increased when lated HIF1a was degraded through a VHL-dependent SUMO-1 was overexpressed (lane 3 versus lane 2 in mechanism. Figure 6A). In contrast, cotransfection of SENP1, but not SENP1 mutant, significantly decreased ubiquitination of HIF1a (lanes 4 and 5 versus 2 in Figure 6A). We asked SUMO Provides an Alternative Signal for HIF1a whether VHL, a well-known E3 ligase for HIF1a ubiquitina- to Bind to VHL tion (Ivan et al., 2001; Jaakkola et al., 2001; Maxwell et al., We further speculated that SUMOylation serves as an al- 1999; Ohh et al., 2000), could also serve as an E3 ligase for ternative signal for HIF1a to bind to VHL during the hyp- SUMOylated HIF1a. To address this possibility, we first oxic condition. The interaction of SUMOylated HIF1a examined hypoxia-induced SUMOylation of HIF1a in and VHL in vivo was examined first by using a HIF1a pro- RCC4 (renal cancer cells with inactivated VHL) and line residue 402/564 mutant (PM), which would decrease RCC4/VHL (VHL-restored RCC4 cells) (Maxwell et al., background binding that resulted from hydroxylation of

590 Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. Figure 6. VHL Is Required for Degrada- tion of SUMOylated HIF1a (A) HIF1a ubiquitination was regulated by SUMO-1 and SENP1. COS-7 cells were co- transfected with indicated plasmids and treated with MG132 (10 mM) for 9 hr before har- vesting. HIF1a was immunoprecipitated with anti-HIF1a (IP) and bound proteins were de- tected by anti-HA (top panel), anti- (sec- ond panel), and anti-HIF1a immunoblotting (third panel) (IB). WCL were immunoblotted (IB) with anti-HA (fourth panel) or anti-Flag (bot- tom panel) antibodies. Asterisk indicates IgG band. (B) RCC4 or RCC4/VHL cells were treated by

hypoxia (1% O2) and/or MG132 (10 mM) for 4 hr as indicated. HIF1a was immunoprecipi- tated with anti-HIF1a antibody from cell lysates. The precipitates were IB with anti- SUMO-1 or -HIF1a antibodies. Asterisk indi- cates IgG band. (C) VHL bound to SUMOylated HIF1a proline mutant (HIF1aPM) in vivo. COS-7 cells were cotransfected with indicated plasmids and treated without or with MG132 (10 mM) for 4 hr before harvesting. VHL was immunopre- cipitated with anti-Flag (IP) from the nuclear fraction of the transfected cells and bound pro- teins were detected by anti-HIF1a (top panel), anti-HA (second panel), or anti-Flag immuno- blotting (third panel) (IB). HIF1a was also immu- noprecipitated with anti-HIF1a (IP) from the nuclear fraction of transfected cells and bound proteins were detected by anti-HA antibody (IB) (fourth panel). WCL were IB with anti- HIF1a (bottom panel). Asterisk indicates IgG band. (D) VHL specifically bound to SUMOylated HIF1a ODD with proline mutation (GST- ODD[344–698] PM) in vitro. GST-ODD(344– 698) PM recombinant protein and SUMOylated GST-ODD(344–698) PM produced by in vitro SUMOylation were incubated with Flag-VHL produced by in vitro translation. After washing and eluting with Flag peptide, the precipitates were immunoblotted with ant-HIF1a (middle panel) or anti-SUMO-1(right panel) antibodies. (E) Mapping of VHL domain that bound to SUMO-1-fused ODD. GST-ODD(344–698) PM and SUMO-1-fused GST-ODD(344–698) PM recombinant proteins were incubated with HA-VHL and its mutant produced by in vitro translation for 2 hr. The precipitates with glutathione-agarose beads were detected by immunoblotting with ant-HA (top and right panel) or anti-HIF1a antibodies (bottom panel). (F) HIF1a degradation induced by SENP1 silencing was VHL dependent. RCC4 or RCC4/VHL cells and SENP1-siRNA stable transfected RCC4 or

RCC4/VHL cells were treated by hypoxia (1% O2) for 4 hr as indicated. Cell lysates were detected by immunoblotting with anti-HIF1a or anti-actin antibodies.

HIF1a. As shown in Figure 6C, SUMOylation of HIF1a PM (Figure 6D, middle panel). These bands were confirmed as was greatly enhanced by expression of SENP1siRNA plus SUMOylated HIF1a by anti-SUMO-1 antibody (Figure 6D, MG132 treatment (lane 4, bottom panel, Figure 6C). SU- right panel). Importantly, unmodified GST-ODD(344-698) MOylated HIF1a from the nuclear fraction was efficiently PM, even though presented in a much larger quantity coprecipitated by VHL (lane 4 in top and second panels, than SUMOylated GST-ODD(344-698) PM, could not be Figure 6C). To directly confirm the binding of VHL and SU- coprecipitated by VHL. We further mapped the SUMO- MOylated HIF1a, we performed an in vitro binding assay. binding domain on VHL protein by using recombinant As shown in Figure 6D, VHL pulled down SUMOylated SUMO-fused GST-ODD(344-698) PM to pull down GST-ODD(344-698) PM, detected by anti-HIF1a antibody in vitro-translated VHL deletion mutants. As shown in

Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. 591 Figure 6E, SUMO-fused GST-ODDPM protein could pull down VHL deletion mutants that contained the b domain. However, SUMO-fused GST-ODDPM protein could not pull down the VHL deletion mutant that only contained the a domain. This was dependent on SUMO-1 because GST-ODDPM could not pull down any VHL deletion mu- tants. Interestingly, b domain is well documented as a sub- strate-binding region when VHL acts as a component of the E3 ligase complex (Ohh et al., 2000; Stebbins et al., 1999). Taken together, these results clearly suggest that SUMOylation of HIF1a could serve as an alternate signal for binding to VHL in the absence of proline hydroxylation. To directly demonstrate the role of SENP1 in VHL-de- pendent degradation of SUMOylated HIF1a, we gener- ated SENP1 siRNA stably transfected RCC4 and RCC4/ VHL cell lines. The expression of endogenous SENP1 was significantly decreased in these two cell lines Figure 7. SENP1 Regulates HIF1a Stability in Hypoxia (Figure S8). Silencing of SENP1 markedly decreased Hypoxia blocks the activity of PHD, preventing hydroxylation of HIF1a HIF1a expression in RCC4/VHL but not in RCC4 cells and its subsequent degradation in a VHL- and ubiquitin-dependent (Figure 6F), confirming that HIF1a turnover promoted by manner. On the other hand, hypoxia induces nuclear translocation inactivation of SENP1 was VHL dependent. and SUMOylation of HIF1a, which provides an alternative signal for VHL- and ubiquitin-dependent degradation. SENP1 stabilizes HIF1a DISCUSSION by removing the alternative VHL-binding signal.

Biological Consequence of SUMOylation HIF1a is hydroxylated at two critical proline residues by and DeSUMOylation a family of oxygen-sensitive , prolyl 4-hydroxy- SUMOylation and deSUMOylation have been shown to lases (PHD) (Bruick and McKnight, 2001; Epstein et al., regulate a large number of biological processes, including 2001; Yu et al., 2001). Proline-hydoxylated HIF1a then transcription, cell signaling, cell-cycle progression, and binds to VHL, a component of the ubiquitin E3 ligase com- cancer pathogenesis (Cheng et al., 2006; Yeh et al., plex that consists of Cul-2, VHL, elongin B, and elongin C 2000). However, many of these studies were carried out (Bruick and McKnight, 2001; Ivan et al., 2001; Jaakkola in cell culture systems overexpressing enzymes involved et al., 2001; Maxwell et al., 1999). Subsequently, HIF1a in SUMOylation and deSUMOylation. A limited number is ubiquitinated and degraded by the proteasome. It was of studies have addressed the physiological importance assumed that during hypoxia, proline hydroxylation oc- of this emerging pathway of biological regulation. For ex- curs inefficiently, thus allowing HIF1a to escape binding ample, knocking out the SUMO conjugation Ubc9 to VHL and proteasomal degradation (Figure 7). Most in mice resulted in embryonic lethality at the early post-im- studies about HIF1a stability primarily focus on the mech- plantation stage (Nacerddine et al., 2005). Ubc9-deficient anism involving the regulation of PHD enzymatic activities cells showed severe defects in nuclear organization in- (Epstein et al., 2001; Gerald et al., 2004; Ivan et al., 2001; cluding condensation and segregation. Jaakkola et al., 2001; Nakayama et al., 2004). However, This study indicates that global disruption of the SUMOy- a few reports have shown that there are other factors lation pathway is lethal in mammals. Consistent with this, that can regulate HIF1a stability in a VHL-independent mice with an incomplete SENP1 knockout die between manner. For example, Liu et al. recently showed that E12.5 and E14.5 (Yamaguchi et al., 2005). RACK1 could replace HSP90’s binding of HIF1a and tar- In the present study, we show that SENP1 plays an es- get the unmodified HIF1a to the elongin C complex for sential role in the deconjugation of SUMOylated HIF1a. ubiquitination and degradation (Liu et al., 2007). There This deconjugation has important biological consequence are also modifications other than hydroxylation that regu- as SENP1/ embryos are severely anemic, which is most late HIF1a stability and activity (Brahimi-Horn et al., 2005). likely the cause of fetal death. This is surprising because in Our SENP1/ embryos demonstrate that a SUMOyla- an overexpression system, both SENP1 and SENP2 have tion and deSUMOylation cycle is involved in the regulation identical substrate specificity. Here, SUMOylated HIF1a is of HIF1a stability during hypoxia. Hypoxia induces nuclear only regulated by SENP1 but not SENP2 in vivo. Thus, translocation (Kallio et al., 1998) and, as we have shown SENPs clearly perform nonredundant biological functions here, SUMOylation of HIF1a, which binds to VHL in a hy- in both MEF cells and animals. droxyl proline-independent manner, leading to ubiquitina- tion and proteasomal degradation (Figure 7). It should be Regulation of HIF1a Stability during Hypoxia noted that SUMOylation is not required for hypoxia-in- HIF1a is mainly regulated at the level of protein stability duced nuclear translocation of HIF1a (X.K. and E.T.H.Y., (Huang et al., 1998; Jiang et al., 1996). During normoxia, unpublished data). We currently do not know how hypoxia

592 Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. induces SUMOylation of HIF1a. This may be due to an in- deleted, SUMOylated HIF1a is degraded, in a VHL- and crease in SUMO E3 ligase activity (X.K. and E.T.H.Y., un- ubiquitin/proteasome-dependent manner, resulting in de- published data). SENP1, which is predominately a nuclear creased Epo production and severe fetal anemia. This protein, is well positioned to regulate the stability of HIF1a study provides evidence for the physiological role of de- in the nucleus by removing SUMO (Gong et al., 2000). SUMOylation as demonstrated in an animal model; our Unmodified HIF1a would escape VHL/proteasome- study also reveals that SUMOylation can target a protein dependent degradation to participate in the regulation for ubiquitination and proteasomal degradation. of hypoxia-responsive genes. When SENP1 is absent, SUMOylated HIF1a will be degraded through a VHL/pro- EXPERIMENTAL PROCEDURES teasome-dependent mechanism (Figure 6). Groulx et al. have proposed that HIF1a is degraded in the Generation of SENP1/ Mice (Groulx and Lee, 2002). However, we do not know whether The SENP1+/ ES cell line XG001 was obtained from BayGenomics. SUMOylated and ubiquitinated HIF1a is also degraded XG001 cells were generated by using a gene trap protocol with the trapping construct pGT1Lxf containing the intron from the - in the cytosol. Further studies are required to resolve 2 gene upstream of the gene encoding the b-galactosidase/neomy- this issue. cin-resistance fusion protein (see http://baygenomics.ucsf.edu). The A previous study showed that overexpression of vector was inserted into intron 8 of the SENP1 locus. A male chimeric SUMO-1 increased HIF1a protein level and its activity mouse was generated from the ES cell line. C57 BL6 mice were ob- not only under hypoxia but also in normoxia condition tained from The Jackson Laboratory. (Bae et al., 2004), leading to the hypothesis that SUMOy- lation stabilizes HIF1a. This model is inconsistent with our Plasmids and Antibodies results demonstrating that SUMOylation leads to the deg- The Epo enhancer (Epo-Luc) and the HIF1a-binding site mutant radation of HIF1a. The contrasting results from the previ- (mEpo-Luc) were provided by Dr. H.M. Sucov. HRE-Luc was provided by Dr. J. Yi. pGal4-VP16 and pGal4-ODD(344–698)-VP16 were pro- ous study may be attributed to the overexpression of vided by Dr. P.J. Ratcliffe. Flag-VHL was provided by Dr. T. Kamitani. SUMO-1 in the presence of SENP1. In the presence of VHL(1–213), VHL(54–213), VHL(63–155), and VHL(156–213) were pro- SENP1, SUMOylated HIF1a will be deconjugated and in- vided by Dr. M. Ohh. HA-SUMO-1, Flag-SENP1, and Flag-SENP1 cat- deed stabilized. The SENP1 knockout mouse has enabled alytic mutant were previously described (1, 2). RGS-HIF1a, RGS- us to uncover an opposite role for SUMOylation in the reg- HIF1aK391R, RGS-HIF1aK477R, RGS-HIF1aSM (K391R, K477R), ulation of HIF1a. RGS-HIF1a PM (P402A, P564A), pET-ODD(344–698)PM(p402A, P564A), and pET-ODD(344–698)PM-SUMO1 were generated using standard cloning procedures and PCR-based mutagenesis. We used SUMOylation Can Target a Protein for antibodies against Flag (M2, Sigma), HA (HA-7, Sigma), Myc (Santa Proteasomal Degradation Cruz), SUMO-1 (Zymed), SUMO-2/3 (gifted from Dr. Mike Matunis), Our results provide direct evidence that SUMOylation can mouse HIF1a (Novus), and human HIF1a (BD). induce degradation of a target protein in a proteasome- dependent manner. It is well accepted that SUMOylation Histopathologic and Immunohistochemistry Analysis can stabilize target proteins, as SUMO can conjugate to Embryos were fixed in 10% buffered formalin (Sigma) and embedded the same lysine sites on target proteins as in ubiquitination in paraffin. Five-micrometer-thick sections were stained with hema- (Hay, 2005). Although ubiquitin-conjugated sites on the toxylin and eosin. For proliferation studies, the sections were stained with Ki67-specific antibodies (Dako). Apoptotic cells were detected HIF1a protein have not been defined precisely, the re- in sections using the TUNEL stain (TACS TdT DAB kit, R&D). ported ubiquitin-conjugated region of HIF1a indicates that at least lysines 391 and 477, two SUMOylation sites Colony-Formation Assays on the HIF1a protein, are not the major sites for ubiquitina- Cells were prepared from the livers of E13.5 embryos in a-MEM tion (Ivan et al., 2001; Masson et al., 2001; Ohh et al., (GIBCO–BRL) and counted in the presence of 3% acetic acid, which 2000). We also found that the level of HIF1a SUMOylation lysed erythrocytes. Cell suspensions were mixed with MethoCult correlated with the level of ubiquitination, suggesting that M3334 to detect BFU-e or MethoCult M3434 to detect CFU-e (Stem- SUMO conjugation of HIF1a is not on the same sites as Cell Technologies). Cells were plated in 35 mm dishes and cultured at ubiquitination. More importantly, we identified an E3 ubiq- 37 C in an atmosphere containing 5% CO2. For the CFU-e assay, ben- zidine-positive CFU-e colonies were scored on day 3. For the BFU-E uitin ligase VHL for SUMOylated HIF1a degradation. It was assay, benzidine-positive BFU-e colonies were scored on day 8. previously shown that PML could be SUMOylated and ubiquitinated when exposed to arsenic trioxide (Lalle- Flow Cytometry mand-Breitenbach et al., 2001). However, it is not known Single-cell suspensions were obtained from E13.5 wild-type and mu- whether SUMOylated PML is required for binding to an tant fetal livers. Cell suspensions were first incubated on ice with rat to allow for ubiquitination and degrada- anti-mouse CD16/CD32 (PharMingen) to block nonspecific binding tion. Currently, we also do not know whether SUMO-me- to Fc receptors. Subsequently, cells were incubated with rat anti- diated binding of substrates to ubiquitin E3 can be gener- mouse PE-conjugated anti-c-kit and anti-CD44 and FITC-conjugated anti-CD34 and anti-Ter-119 (all from PharMingen). Appropriate isotype alized to more SUMOylated substrates. control antibodies were used. Cell-surface expression of different In conclusion, our results support a model in which SU- markers was analyzed in a Becton Dickinson FACScan using Cell- MOylated HIF1a is unstable but can be stabilized when Quest software. For the TUNEL assay, cells were stained using a SUMO is removed by SENP1 (Figure 7). When SENP1 is TUNEL kit from Roche and then analyzed by flow cytometry.

Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. 593 RNA Interference Di Bacco, A., Ouyang, J., Lee, H.Y., Catic, A., Ploegh, H., and Gill, G. Two 21 nucleotide SENP1 siRNAs (si-1: AACTACATCTTCGTG (2006). The SUMO-specific protease SENP5 is required for cell divi- TACCTC; si-2: CTAAACCATCTGAATTGGCTC) were synthesized sion. Mol. Cell. Biol. 26, 4489–4498. (Dharmacon). The same sequence of the si-1-inverted orientation Ebert, B.L., and Bunn, H.F. (1999). Regulation of the erythropoietin was used as a nonspecific siRNA control. The SENP1 and nonspecific gene. Blood 94, 1864–1877. siRNA oligos were inserted into a pSuppressorNeo vector (IMGENEX Epstein, A.C., Gleadle, J.M., McNeill, L.A., Hewitson, K.S., O’Rourke, Corporation) according to manufacturer’s instructions. Hep 3B cells J., Mole, D.R., Mukherji, M., Metzen, E., Wilson, M.I., Dhanda, A., were transfected with the siRNA plasmid using lipofectamine 2000 (In- et al. (2001). C. elegans EGL-9 and mammalian homologs define a fam- vitrogen). RCC4 and RCC4/VHL (purchased from ECACC) were in- ily of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107, fected by retrovirus-based SENP1si-1 virus particles produced from 43–54. pSuppressorNeo-SENP1si-1 and selected by G418. Silencing effi- ciency of the siRNA was confirmed by performing real-time PCR (for Gerald, D., Berra, E., Frapart, Y.M., Chan, D.A., Giaccia, A.J., Mansuy, Hep 3B) or RT-PCR (for RCC4 and RCC4/VHL) to examine SENP1 ex- D., Pouyssegur, J., Yaniv, M., and Mechta-Grigoriou, F. (2004). JunD pression. TaqMan Master Mix Reagents (Applied Biosystems) were reduces tumor by protecting cells from oxidative stress. utilized for quantitative real-time PCR (QRT-PCR) reaction. The Taq- Cell 118, 781–794. Man ABI PRISM 7000 Sequence Detector System (PE Applied Biosys- Giaccia, A.J., Simon, M.C., and Johnson, R. (2004). The biology of hyp- tems) was used for the analysis. Primers for SENP1 (forward: 50- oxia: the role of oxygen sensing in development, normal function, and TTGGCCA GAGTGCAAATGG-30; reverse: 50-TCGGCTGTTTCTTGA disease. Genes Dev. 18, 2183–2194. TTTTTGTAA-30) and the housekeeping 18S rRNA (ABI) were utilized. Gill, G. (2004). SUMO and ubiquitin in the nucleus: different functions, similar mechanisms? Genes Dev. 18, 2046–2059.

In Vitro SUMOylation Assay Godin, I., and Cumano, A. (2002). The hare and the tortoise: an embry- In vitro SUMOylation kit was purchased from LAE Biotech International onic haematopoietic race. Nat. Rev. Immunol. 2, 593–604. (Rockville, MD). The reaction was carried out at 37C for 1 hr with the Gong, L., and Yeh, E.T. (2006). Characterization of a family of nucleolar mixture including E1 (150 ng), E2 (5 mg), SUMO-1 (5 mg), ATP (2 mM), SUMO-specific proteases with preference for SUMO-2 or SUMO-3. J. and GST-ODD(344–698)PM (300 ng). Biol. Chem. 281, 15869–15877. Gong, L., Millas, S., Maul, G.G., and Yeh, E.T. (2000). Differential reg- ulation of sentrinized proteins by a novel sentrin-specific protease. J. Supplemental Data Biol. Chem. 275, 3355–3359. Supplemental Data include Supplemental Experimental Procedures and nine figures and can be found with this article online at http:// Groulx, I., and Lee, S. (2002). Oxygen-dependent ubiquitination and www.cell.com/cgi/content/full/131/3/584/DC1/. degradation of hypoxia-inducible factor requires nuclear-cytoplasmic trfficking of von Hippel-Lindau tumor suppressor protein. Mol. Cell. Biol. 22, 5319–5336. ACKNOWLEDGMENTS Hay, R.T. (2005). SUMO: a history of modification. Mol. Cell 18, 1–12. Huang, L.E., Gu, J., Schau, M., and Bunn, H.F. (1998). Regulation of We thank J. Ni, J. Ren, S. Cao, and L. Zhao for expert technical assis- hypoxia-inducible factor 1alpha is mediated by an O2-dependent deg- tance. This work was supported in part by NIH R01 CA 80089 radation domain via the ubiquitin-proteasome pathway. Proc. Natl. (E.T.H.Y), Department of Defense-CDMRP PC040121 (E.T.H.Y.), and Acad. Sci. USA 95, 7987–7992. CA-16672 (MDACC). Ivan, M., Kondo, K., Yang, H., Kim, W., Valiando, J., Ohh, M., Salic, A., Received: April 16, 2007 Asara, J.M., Lane, W.S., and Kaelin, W.G., Jr. (2001). HIFalpha tar- Revised: July 12, 2007 geted for VHL-mediated destruction by proline hydroxylation: implica- Accepted: August 27, 2007 tions for O2 sensing. Science 292, 464–468. Published: November 1, 2007 Iyer, N.V., Kotch, L.E., Agani, F., Leung, S.W., Laughner, E., Wenger, R.H., Gassmann, M., Gearhart, J.D., Lawler, A.M., Yu, A.Y., and Se- menza, G.L. (1998). Cellular and developmental control of O2 homeo- REFERENCES stasis by hypoxia-inducible factor 1 alpha. Genes Dev. 12, 149–162.

Bae, S.H., Jeong, J.W., Park, J.A., Kim, S.H., Bae, M.K., Choi, S.J., and Jaakkola, P., Mole, D.R., Tian, Y.M., Wilson, M.I., Gielbert, J., Gaskell, Kim, K.W. (2004). Sumoylation increases HIF-1alpha stability and its S.J., Kriegsheim, A., Hebestreit, H.F., Mukherji, M., Schofield, C.J., transcriptional activity. Biochem. Biophys. Res. Commun. 324, 394– et al. (2001). Targeting of HIF-alpha to the von Hippel-Lindau ubiquity- 400. lation complex by O2-regulated prolyl hydroxylation. Science 292, 468–472. Brahimi-Horn, C., Mazure, N., and Pouyssegur, J. (2005). Signalling via Jiang, B.H., Semenza, G.L., Bauer, C., and Marti, H.H. (1996). Hyp- the hypoxia-inducible factor-1alpha requires multiple posttranslational oxia-inducible factor 1 levels vary exponentially over a physiologically modifications. Cell. Signal. 17, 1–9. relevant range of O2 tension. Am. J. Physiol. 271, C1172–C1180. Bruick, R.K., and McKnight, S.L. (2001). A conserved family of prolyl-4- Kallio, P.J., Okamoto, K., O’Brien, S., Carrero, P., Makino, Y., Tanaka, hydroxylases that modify HIF. Science 294, 1337–1340. H., and Poellinger, L. (1998). Signal transduction in hypoxic cells: in- Cheng, J., Wang, D., Wang, Z., and Yeh, E.T. (2004). SENP1 enhances ducible nuclear translocation and recruitment of the CBP/p300 coac- -dependent transcription through desumoylation of tivator by the hypoxia-inducible factor-1alpha. EMBO J. 17, 6573– histone deacetylase 1. Mol. Cell. Biol. 24, 6021–6028. 6586. Cheng, J., Perkins, N.D., and Yeh, E.T. (2005). Differential regulation of Kondo, M., Wagers, A.J., Manz, M.G., Prohaska, S.S., Scherer, D.C., c-Jun-dependent transcription by SUMO-specific proteases. J. Biol. Beilhack, G.F., Shizuru, J.A., and Weissman, I.L. (2003). Biology of he- Chem. 280, 14492–14498. matopoietic stem cells and progenitors: implications for clinical appli- Cheng, J., Bawa, T., Lee, P., Gong, L., and Yeh, E.T. (2006). Role of de- cation. Annu. Rev. Immunol. 21, 759–806. sumoylation in the development of cancer. Neoplasia 8, 667– Lallemand-Breitenbach, V., Zhu, J., Puvion, F., Koken, M., Honore, N., 676. Doubeikovsky, A., Duprez, E., Pandolfi, P.P., Puvion, E., Freemont, P.,

594 Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. and de The, H. (2001). Role of promyelocytic leukemia (PML) sumola- Parganas, E., Wang, D., Stravopodis, D., Topham, D.J., Marine, J.C., tion in nuclear body formation, 11S proteasome recruitment, and Teglund, S., Vanin, E.F., Bodner, S., Colamonici, O.R., van Deursen, As2O3-induced PML or PML/ alpha degradation. J.M., et al. (1998). Jak2 is essential for signaling through a variety of cy- J. Exp. Med. 193, 1361–1371. tokine receptors. Cell 93, 385–395.

Liu, Y.V., Baek, J.H., Zhang, H., Diez, R., Cole, R.N., and Semenza, Ryan, H.E., Lo, J., and Johnson, R.S. (1998). HIF-1 alpha is required for G.L. (2007). RACK1 competes with HSP90 for binding to HIF-1alpha solid tumor formation and embryonic vascularization. EMBO J. 17, and is required for O(2)-independent and HSP90 inhibitor-induced 3005–3015. degradation of HIF-1alpha. Mol. Cell 25, 207–217. Masson, N., Willam, C., Maxwell, P.H., Pugh, C.W., and Ratcliffe, P.J. Socolovsky, M., Fallon, A.E., Wang, S., Brugnara, C., and Lodish, H.F. (2001). Independent function of two destruction domains in hypoxia- (1999). Fetal anemia and apoptosis of red cell progenitors in Stat5a/ inducible factor-alpha chains activated by prolyl hydroxylation. 5b/ mice: a direct role for Stat5 in Bcl-X(L) induction. Cell 98, 181– EMBO J. 20, 5197–5206. 191. Maxwell, P.H., Wiesener, M.S., Chang, G.W., Clifford, S.C., Vaux, E.C., Stebbins, C.E., Kaelin, W.G., Jr., and Pavletich, N.P. (1999). Structure Cockman, M.E., Wykoff, C.C., Pugh, C.W., Maher, E.R., and Ratcliffe, of the VHL-ElonginC-ElonginB complex: implications for VHL tumor P.J. (1999). The tumour suppressor protein VHL targets hypoxia-in- suppressor function. Science 284, 455–461. ducible factors for oxygen-dependent . Nature 399, 271– Wang, G.L., Jiang, B.H., Rue, E.A., and Semenza, G.L. (1995). Hyp- 275. oxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer Nacerddine, K., Lehembre, F., Bhaumik, M., Artus, J., Cohen-Tan- regulated by cellular O2 tension. Proc. Natl. Acad. Sci. USA 92, noudji, M., Babinet, C., Pandolfi, P.P., and Dejean, A. (2005). The 5510–5514. SUMO pathway is essential for nuclear integrity and chromosome seg- regation in mice. Dev. Cell 9, 769–779. Wu, H., Liu, X., Jaenisch, R., and Lodish, H.F. (1995). Generation of Nakayama, K., Frew, I.J., Hagensen, M., Skals, M., Habelhah, H., committed erythroid BFU-E and CFU-E progenitors does not require Bhoumik, A., Kadoya, T., Erdjument-Bromage, H., Tempst, P., Frap- erythropoietin or the erythropoietin receptor. Cell 83, 59–67. pell, P.B., et al. (2004). Siah2 regulates stability of prolyl-hydroxylases, Yamaguchi, T., Sharma, P., Athanasiou, M., Kumar, A., Yamada, S., controls HIF1alpha abundance, and modulates physiological re- and Kuehn, M.R. (2005). Mutation of SENP1/SuPr-2 reveals an essen- sponses to hypoxia. Cell 117, 941–952. tial role for desumoylation in mouse development. Mol. Cell. Biol. 25, Neubauer, H., Cumano, A., Muller, M., Wu, H., Huffstadt, U., and 5171–5182. Pfeffer, K. (1998). Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis. Cell 93, 397–409. Yeh, E.T., Gong, L., and Kamitani, T. (2000). Ubiquitin-like proteins: new wines in new bottles. Gene 248, 1–14. Ohh, M., Park, C.W., Ivan, M., Hoffman, M.A., Kim, T.Y., Huang, L.E., Pavletich, N., Chau, V., and Kaelin, W.G. (2000). Ubiquitination of hyp- Yu, F., White, S.B., Zhao, Q., and Lee, F.S. (2001). HIF-1alpha binding oxia-inducible factor requires direct binding to the beta-domain of the to VHL is regulated by stimulus-sensitive proline hydroxylation. Proc. von Hippel-Lindau protein. Nat. Cell Biol. 2, 423–427. Natl. Acad. Sci. USA 98, 9630–9635.

Cell 131, 584–595, November 2, 2007 ª2007 Elsevier Inc. 595