Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Accepted manuscripts
    • Latest complete issue
    • Issue archive
    • Archive by article type
    • Special issues
    • Subject collections
    • Cell Scientists to Watch
    • First Person
    • Sign up for alerts
  • About us
    • About JCS
    • Editors and Board
    • Editor biographies
    • Travelling Fellowships
    • Grants and funding
    • Journal Meetings
    • Workshops
    • The Company of Biologists
    • Journal news
  • For authors
    • Submit a manuscript
    • Aims and scope
    • Presubmission enquiries
    • Fast-track manuscripts
    • Article types
    • Manuscript preparation
    • Cover suggestions
    • Editorial process
    • Promoting your paper
    • Open Access
    • JCS Prize
    • Manuscript transfer network
    • Biology Open transfer
  • Journal info
    • Journal policies
    • Rights and permissions
    • Media policies
    • Reviewer guide
    • Sign up for alerts
  • Contacts
    • Contact JCS
    • Subscriptions
    • Advertising
    • Feedback
  • COB
    • About The Company of Biologists
    • Development
    • Journal of Cell Science
    • Journal of Experimental Biology
    • Disease Models & Mechanisms
    • Biology Open

User menu

  • Log in

Search

  • Advanced search
Journal of Cell Science
  • COB
    • About The Company of Biologists
    • Development
    • Journal of Cell Science
    • Journal of Experimental Biology
    • Disease Models & Mechanisms
    • Biology Open

supporting biologistsinspiring biology

Journal of Cell Science

  • Log in
Advanced search

RSS   Twitter  Facebook   YouTube  

  • Home
  • Articles
    • Accepted manuscripts
    • Latest complete issue
    • Issue archive
    • Archive by article type
    • Special issues
    • Subject collections
    • Cell Scientists to Watch
    • First Person
    • Sign up for alerts
  • About us
    • About JCS
    • Editors and Board
    • Editor biographies
    • Travelling Fellowships
    • Grants and funding
    • Journal Meetings
    • Workshops
    • The Company of Biologists
    • Journal news
  • For authors
    • Submit a manuscript
    • Aims and scope
    • Presubmission enquiries
    • Fast-track manuscripts
    • Article types
    • Manuscript preparation
    • Cover suggestions
    • Editorial process
    • Promoting your paper
    • Open Access
    • JCS Prize
    • Manuscript transfer network
    • Biology Open transfer
  • Journal info
    • Journal policies
    • Rights and permissions
    • Media policies
    • Reviewer guide
    • Sign up for alerts
  • Contacts
    • Contact JCS
    • Subscriptions
    • Advertising
    • Feedback
Research Article
Mechanisms of the HRSL3 tumor suppressor function in ovarian carcinoma cells
Irina Nazarenko, Reinhold Schäfer, Christine Sers
Journal of Cell Science 2007 120: 1393-1404; doi: 10.1242/jcs.000018
Irina Nazarenko
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Reinhold Schäfer
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christine Sers
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & tables
  • Supp info
  • Info & metrics
  • PDF
Loading

Summary

HRSL3 (also known as H-REV107-1) belongs to a class II tumor suppressor gene family and is downregulated in several human tumors including ovarian carcinomas. To unravel the mechanism of HRSL3 tumor suppressor action, we performed a yeast two-hybrid screen and identified the α-isoform of the regulatory subunit A of protein phosphatase 2A (PR65α) as a new interaction partner of HRSL3. Interaction between HRSL3 and PR65α was confirmed in vitro and by co-immunoprecipitation in mammalian cells. We demonstrate that HRSL3 binds to the endogenous PR65α, thereby partially sequestering the catalytic subunit PR36 from the PR65 protein complex, and inhibiting PP2A catalytic activity. Furthermore, binding of HRSL3 to PR65 induces apoptosis in ovarian carcinoma cells in a caspase-dependent manner. Using several mutant HRSL3 constructs, we identified the N-terminal proline-rich region within the HRSL3 protein as the domain that is relevant for both binding of PR65α and induction of programmed cell death. This suggests that the negative impact of HRSL3 onto PP2A activity is important for the HRSL3 pro-apoptotic function and indicates a role of PP2A in survival of human ovarian carcinomas. The analysis of distinct PP2A target molecules revealed PKCζ as being involved in HRSL3 action. These data implicate HRSL3 as a signaling regulatory molecule, which is functionally involved in the oncogenic network mediating growth and survival of ovarian cancer cells.

  • Tumor suppressor
  • HRSL3
  • PP2A
  • Apoptosis

Introduction

The inactivation of tumor suppressors is an essential step during tumorigenesis. The canonical, class I tumor suppressor genes, such as p53 or Rb, are targeted by genetic alterations, which abrogate the activity of their gene products in transformed cells. Alternatively, tumor suppressors can be inhibited by reversible mechanisms, while their genomic structure remains intact. These genes, classified as class II tumor suppressors (Sager, 1997), are particularly important in view of their potential relevance for cancer therapy. Expression of these genes can be recovered in tumor cells by inhibition of signaling pathways or de-methylating agents, and the genes can expose their tumor suppressing properties.

Recently, a novel gene family of class II tumor suppressors, designated as the H-REV107-like family, was postulated (Hughes and Stanway, 2000). Five of the H-REV107-like proteins are found in the human genome and are deregulated in distinct tumors and tumor-derived cell lines. HRASL3 encodes the HRSL3 protein (also known as H-REV107-1) and is expressed in most normal tissues but is lost in human tumors derived from breast, ovary, kidney and testicular germ cells (Husmann et al., 1998; Siegrist et al., 2001). The HRSL3 protein suppresses colony formation in RAS-transformed fibroblasts and attenuates tumor growth in nude mice (Sers et al., 1997). In ovarian carcinoma cells, overexpression of human HRSL3 results in cell death (Sers et al., 2002), and loss of the IRF-1 transcription factor was demonstrated to be responsible for the reversible downregulation of HRSL3 (Sers et al., 2002). The closely related RARRES3 (also known as TIG3) gene is suppressed in basal and squamous cell carcinomas of the skin, and in breast and gastric cancer cell lines (DiSepio et al., 1998; Huang et al., 2000). RARRES3 impairs the function of several mitogen-activated and extracellular signal-regulated protein kinases in human cervical cancer cells (Huang et al., 2002) and was recently shown to suppress RAS activation (Tsai et al., 2006). Furthermore, the protein induces differentiation in human foreskin keratinocytes (Sturniolo et al., 2003). The recently identified HRASLS gene is suppressed in human gastric cancers (Kaneda et al., 2002), but its involvement in growth regulation was not defined.

The H-REV107-like proteins share four highly conserved domains, a proline-rich motif located at the N terminus, two conserved boxes designated according to their amino acid sequences as the HWAIY-box and the NCE-box, and a hydrophobic C-terminal putative transmembrane domain. This structural conservation among members of the H-REV107-like protein family suggests a similarity of their functions. The five H-REV107-like proteins are structurally most related to lecithine retinol acyltransferase (LRAT), an enzyme involved in the metabolism of retinoic acid (MacDonald and Ong, 1988). Although none of the H-REV107-like proteins was reported to possess an enzymatic activity, phylogenetic analysis showed that H-REV107-like proteins and LRAT belong to the NlpC/P60 superfamily, which consists mostly of prokaryotic peptidases (Anantharaman and Aravind, 2003). NlpC/P60 proteins share the histidine (His) and cysteine (Cys) residues that correspond to the histidine in the HWAIY and the central cysteine in the NCE boxes of the H-REV107-like and LRAT proteins. Only in the LRAT protein, were the conserved histidine and cysteine residues demonstrated to influence LRAT catalytic activity (Xue and Rando, 2004), whereas the role of the N-terminal domain was not investigated in this protein. By contrast, the C-terminus was shown to be responsible for the intracellular localization and growth suppressive function of HRSL3 and RARRES3 (Deucher et al., 2000; Sers et al., 1997).

In the current work, we aimed to define the mechanism of HRSL3 suppressive action and to further investigate the functional relevance of the highly conserved domains. We performed a yeast two-hybrid interaction trap and identified the regulatory subunit Aα of the protein phosphatase 2A (PR65α). We defined HRSL3 domains responsible for the interaction, and investigated the effect of the interaction of HRSL3 and PR65α on the protein phosphatase 2A (PP2A) catalytic activity.

Results

HRSL3 directly binds PR65α, the regulatory subunit A of PP2A, and its N-terminal proline-rich region plays a major role in this interaction

To examine the mechanism by which HRSL3 suppresses cellular growth and induces apoptosis, we accomplished a search for its potential interacting proteins using a LexA-based yeast two-hybrid system. First, we generated a fusion construct containing the DNA-binding domain of the LexA protein and a truncated HRSL3 protein (HREV1dC), lacking 29 amino acids, which comprise its membrane-binding domain. The truncated form of HRSL3 was used because in a library screen with the full-length protein none of the clones survived, suggesting that the membrane-binding domain inhibits the transport of the recombinant protein into the nucleus. Using the truncated HREV1dC fusion construct, we screened a human kidney cDNA library, and successfully isolated several positive clones. One of the clones contained the PPP2R1A gene, encoding PR65α the regulatory subunit Aα of PP2A, which is a major serine/threonine phosphatase in eukaryotic cells (Millward et al., 1999). To confirm the interaction between the truncated HRSL3 (HREV1dC) and PR65α, we first performed co-immunoprecipitation of HA-fused HREV1dC (HREV1dCHA) and V5-fused PR65α (PR65αV5) in COS-7 cells, transiently transfected with the corresponding expression plasmids. Protein complexes were immunoprecipitated either with an anti-V5 or with an anti-HA antibody. Using the anti-V5 antibody for immunoprecipitation, we observed the HREV1dCHA protein only in the presence of PR65αV5 (Fig. 1A, left panel, top). As a control, western blotting with an anti-V5 antibody was used (Fig. 1A, left panel, bottom). The reverse experiment, precipitation with an anti-HA antibody, again confirmed the interaction of PR65αV5 with the HREV1dCHA (Fig. 1A, right panel). These results demonstrated that overexpressed HREV1dCHA and PR65αV5 form a complex in mammalian cells. In a GST pull-down assay we used a GST-HREV1dC recombinant construct containing the truncated HRSL3 protein fused to glutathione and coupled to glutathione-Sepharose. With this approach, we were able to recover PR65α protein from cell lysates thus confirming a direct interaction between the HRSL3 and PR65α proteins (Fig. 1B).

Next, we aimed to determine the domains of HRSL3 governing the interaction with PR65α. For that purpose we designed HRSL3 mutants defective in the conserved domains typical for H-REV107-like proteins (Fig. 1C). The mutant AWAIdCHA, carries a substitution of His23 to Ala within the HWAIY domain. We also modified the NCE domain and generated the NSEdCHA mutant, which involved a one-amino acid exchange of Cys112 to Ser. Additionally, we constructed the dNdCHA mutant, which carries a deletion of 10 N-terminal amino acids including the proline-rich region (Fig. 1C). The HREV1dCHA, AWAIdCHA, NSEdCHA and dNdCHA expression vectors were transiently cotransfected with PR65αV5 into COS-7 cells. Protein interactions were assessed by immunoprecipitation with an anti-V5 antibody. We detected similar amounts of the HREV1dCHA, the AWAIdCHA, and the NSEdCHA proteins in the PR65αV5 immunocomplex. However, the dNdCHA deletion mutant, lacking 10 N-terminal amino acids, did not interact with PR65αV5 (Fig. 1D, left panel, top). Re-probing of this immunoblot with an anti-V5 antibody demonstrated efficient immunoprecipitation of PR65αV5 in all samples (Fig. 1D, left panel, bottom). The reverse experiment, immunoprecipitation with an anti-HA antibody confirmed these results (Fig. 1D, right panel, top). Thus, deletion of the N-terminal region of HRSL3 abrogated its binding to PR65αV5, indicating that this region plays a major role in the interaction between HRSL3 and the regulatory subunit A of PP2A (PR65α).

The endogenous regulatory (PR65) but not the catalytic subunit (PR36) of PP2A is an interaction partner of HRSL3

The interaction between HRSL3 and PR65α suggested that HRSL3 might be a novel component of the PP2A complex. To prove this hypothesis, we aimed to examine the interaction between endogenous HRSL3 and PR65α and tested a number of cell lines for expression of the HRSL3 and the PR65α proteins (Fig. 2A). The regulatory subunit PR65 tightly associates with the catalytic subunit PR36 forming a “core dimer” (Millward et al., 1999), therefore, we also analyzed expression of PR36α. PR65α and PR36α were equally expressed in all cell lines analyzed, however, consistent with our earlier observations (Husmann et al., 1998; Sers et al., 2002), no HRSL3 protein was detected in several rat and human tumor cell lines and in COS-7 cells (Fig. 2A). The absence of the HRSL3 protein in cultured cells excluded investigation of an interaction between HRSL3 and PR65 under physiological conditions. Therefore, for further experiments we used OVCAR-3 cells, an ovarian carcinoma cell line, in which we had demonstrated previously a suppressive function of HRSL3 (Sers et al., 2002) upon transient overexpression of the HRSL3 protein.

We isolated the endogenous PR65α immunocomplex from OVCAR-3 cells using a PR65α-specific antibody bound to an AminoLink-activated coupling gel (Pierce Biotechnology Inc. IL). As an isotype control, we applied the same protein extracts to mouse IgG2a (Fig. 2B). Analysis of the recovered protein complexes revealed the presence of the endogenous regulatory subunit PR65α and the overexpressed HRSL3 protein (Fig. 2B), clearly demonstrating interaction between HRSL3 and endogenous PP2A.

For further evaluation of HRSL3 binding to the endogenous PP2A complex in OVCAR-3 cells, we performed co-immunoprecipitation using transfected full-length HRSL3 (HREV1FL) and the interaction-deficient mutant (dNdC) and tested their binding to the regulatory subunit PR65α (Fig. 2C, left panel) and to the catalytic subunit PR36α of PP2A (Fig. 2C, right panel). Whereas binding of full-length HRSL3 to PR65α was confirmed (Fig. 2C, left panel, bottom), neither wild-type HRSL3 nor the interaction deficient mutant were detected in the PR36 protein complex (Fig. 2C, right panel, bottom). In addition, we examined whether the presence of the HRSL3 protein might influence the amount of the catalytic subunit PR36 and the regulatory subunit B56 precipitated within the PR65α immunocomplex. No difference in the relative amounts of the regulatory B subunit, B56 were observed in immunocomplexes containing either wild-type HRSL3 or its interaction-deficient mutant dNdC (Fig. 2C, left panel, middle). By contrast, a considerably lower amount of the catalytic subunit PR36 was detected in the presence of full-length HRSL3 in the PR65 immunocomplex as compared to the control (Fig. 2C, left panel, asterisk). These results indicate that HRSL3 binds specifically to the regulatory, but not to the catalytic subunit of PP2A and suggest that the interaction between PR65 and HRSL3 might displace PR36 but not B56 from the HRSL3-PR65α complex.

  Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

The N-terminus of the HRSL3 protein (HREV1) is important for the binding of HRSL3 and PR65. (A) Left panel: proteins were immunoprecipitated with an anti-V5 antibody, and subsequently detected with anti-HA (top) and anti-V5 (bottom) antibodies. Lanes (from the left): 1, total cell lysate; 2, immunoprecipitation from cellular extracts harboring both proteins; 3, 4, immunoprecipitation from cellular extracts harboring PR65αV5 or HREV1dCHA, respectively. Right panel: proteins were immunoprecipitated with an anti-HA antibody and subsequently detected with anti-V5 (top) antibody. The efficiency of the immunoprecipitation was controlled using an anti-HA antibody (bottom). Lanes (from the left): 1, immunoprecipitation from cellular extracts harboring both proteins; 2, 3, immunoprecipitation from cellular extracts harboring PR65αV5 or HREV1dCHA, respectively. (B) To prove whether PR65 directly interacts with HRSL3, cleared COS-7 cell lysates were incubated with either GST or GST-HREV1dC proteins coupled to GST-Sepharose. Relative amounts of the proteins were analyzed using western blotting against PR65 and GST antibodies. (C) The HREV1FL expression construct carries wild-type full-length HRASL3 cDNA. Four domains of the HRSL3 protein are indicated: a proline-rich domain at the N terminus of the protein (PD), a HWAIY domain containing a conserved histidine (His23), an NCE domain containing a conserved cystein (Cys112), and a membrane binding domain (MBD). In the HREV1dCHA expression construct, 29 C-terminal amino acids are substituted by the HA-epitope. AWAIdCHA carries a mutation His23→Ala; NSEdCHA carries a mutation Cys112→Ser; dNdCHA harbors a deletion of the N-terminal proline-rich region. (D) COS-7 cells were cotransfected with the following expression plasmids: PR65αV5 and HREV1dCHA, PR65αV5 and HREV1-AWAIYdCHA, PR65αV5 and HREV1-NSEdCHA, and PR65αV5 and HREV1-dNdCHA. As negative controls, plasmids containing HA or V5 epitopes only were transfected (–). (Left panel) Top: proteins were immunoprecipitated with an anti-V5 antibody and subsequently analyzed with an anti-HA antibody. HREV1dCHA, HREV1-AWAIdCHA and HREV1-NSEdCHA associate with PR65αV5, whereas the HREV1-dNdCHA deletion mutant is unable to interact with PR65αV5. Bottom: incubation with an anti-V5 antibody demonstrates equal amounts of the immunoprecipitated PR65αV5 protein. (Right panel) Top: proteins were immunoprecipitated with an anti-HA antibody and subsequently analyzed with an anti-V5 antibody. The PR65αV5 protein was obtained in a complex with HREV1dCHA, HREV1-AWAIdCHA and HREV1-NSEdCHA. By contrast, no PR65αV5 was detected in the immunoprecipitated HREV1-dNdC complex. Bottom: immunoprecipitation was controlled using an anti-HA antibody and demonstrates equal amounts of the HA-tagged HRSL3 proteins.

  Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

HRSL3 (HREV1) interacts with the endogenous regulatory, but not the catalytic, subunits of PP2A. (A) Protein extracts from several cell lines were analyzed by immunoblotting against an HRSL3-specific antibody described previously as H-REV107-1. As a control, COS-7 cells transiently transfected with an HRASL3 expression plasmid, were used. PR36α and PR65α were detected with appropriate antibodies. Actin was used as a loading control. (B) The PR65α immunocomplex was recovered from OVCAR-3 cells transiently transfected with an HRASL3 expression plasmid (HREV1FL). Immunoprecipitated proteins (IP), and protein extracts used for the immunoprecipitation (Input) were subjected to SDS-PAGE and western blot analysis. Presence of the regulatory PR65α (top) and the catalytic PR36α (middle) subunits was demonstrated by immunoblotting with appropriate antibodies. Application of an anti-HRSL3 specific antibody (bottom) revealed the presence of the HRSL3 protein in the PR65α protein complex (second lane) but not in the control immunoprecipitation (third lane), confirming the interaction between HRSL3 and endogenous PR65α. (C) OVCAR-3 cells were transiently transfected with HREV1FL, dNdC and pcDNA3 as a control. Forty-eight hours post-transfection immunoprecipitation was performed with a PR65α-specific antibody (left panel) and with a PR36α,β-specific antibody (right panel). Presence of the HRSL3 full-length and truncated proteins, PR65α,β, PR36α,β and B56 in the protein extracts used for immunoprecipitation (Input) and in the precipitated protein complexes was analyzed using western blotting with the appropriate antibodies. *Decreased amount of the PR36 proteins in the PR65 protein complex in the presence of HRSL3 was semiquantitatively evaluated (see bar chart below) using ImageJ freeware. The ratio between the amounts of immunoprecipitated PR36 and the light chain of the PR65 antibody is shown on top of the bars. (D) OVCAR-3 cells were transiently transfected with HREV1FL-V5 and incubated with PR65, and V5-specific antibodies. Then the cells were stained with DAPI to visualize nuclei (blue), with Alexa Fluor 488 and Alexa Fluor 594 secondary antibodies to visualize the distribution of the PR65α (red) and the HRSL3-V5 (green) proteins, respectively using confocal microscopy. (Merge) Yellow color indicates a colocalization of the HRSL3 and the PR65α proteins (overlay of red and green) in the cytoplasm. (E) OVCAR-3 cells were transiently transfected with HREV1FL and incubated with PR36, and HRSL3-specific antibodies. Then the cells were stained with DAPI to visualize nuclei (blue), with Alexa Fluor 488 and Alexa Fluor 594 secondary antibodies to visualize distribution of the PR36 (green) and the HRSL3 (red) proteins, respectively using confocal microscopy. (Merge) Absence of yellow color indicates no colocalization of the HRSL3 and the PR36 protein in the cells.

We also examined the intracellular distribution of the regulatory (PR65α) and the catalytic (PR36) subunits of PP2A and investigated a potential colocalization with the overexpressed full-length HRSL3 protein (HREV1FL or HREV1FL-V5). In OVCAR-3 cells, PR65α and HRSL3 were preferably localized in the cytoplasm where both proteins are co-distributed (Fig. 2D, merge). Consistently with our previous observations (Sers et al., 2002), HRSL3 overexpression correlated with the development of an apoptotic morphology of the nuclei (Fig. 2D, DAPI). Interestingly, we could not detect a co-distribution of HRSL3 and PR36 (Fig. 2E, merge), further supporting our co-immunoprecipitation data, which suggest that the HRSL3-PR65α complex might not contain PR36.

The N-terminal domain of HRSL3 is required for inhibition of the PP2A catalytic activity

Since HRSL3 interacts with the PP2A regulatory subunit but not with the catalytic subunit, we tested if the catalytic activity of intracellular PP2A would change in the presence of HRSL3 (Fig. 3A). We overexpressed full-length HRSL3 and the interaction-deficient mutant dNdC in OVCAR-3 cells. As controls, we analyzed cells transfected with pcDNA3, and cells treated with the natural PP2A inhibitor okadaic acid (OA) (Holmes et al., 1990). To achieve effective inhibition of PP2A in cell culture, we used 10 nM OA (Fig. 4A) (Favre et al., 1997). At this concentration OA also inhibits several other members of the serine/threonine protein phosphatase family (Bastians and Ponstingl, 1996; Brewis et al., 1993; Cohen et al., 1996; Sasaki et al., 1994), therefore we measured phosphatase activity of the immunoprecipitated PR65 protein complexes (Fig. 3A). In parallel, we controlled the amounts of the precipitated phosphatase and HRSL3 (Fig. 3B). As shown in Fig. 3A, the presence of HRSL3 led to a 40% inhibition of PP2A activity, whereas the interaction-deficient mutant dNdC did not inhibit PP2A, suggesting that the interaction between HRSL3 and PR65α might be important for the diminished catalytic activity of PP2A.

The observed reduction of the PR36 catalytic subunit within the PP2A complex after HRSL3 transfection might be one reason for a lower PP2A activity. However, other mechanisms might contribute to an HRSL3-mediated suppression of PP2A activity. For instance, the intracellular localization of PP2A plays a role in the capacity of PP2A to dephosphorylate particular substrates (Janssens et al., 2005; Janssens and Goris, 2001; Zuluaga et al., 2007). To investigate this possibility, we examined the subcellular distribution of the PP2A regulatory and catalytic subunits in OVCAR-3 cells either treated with OA or transfected with wild-type HRSL3 and the interaction-deficient mutant dNdC. Only moderate inconsistent changes in the protein levels were observed in different subcellular fractions after treatment or HRSL3 overexpression (see Fig. S1 in supplementary material), suggesting that HRSL3 regulates PP2A activity by other mechanisms.

Suppression of PP2A activity by HRSL3 is sufficient to induce apoptosis in ovarian carcinoma cells

In our previous investigation, we had detected loss of HRSL3 from human ovarian carcinomas and demonstrated that overexpression of the HRSL3 protein induced apoptosis in ovarian cancer cells (Sers et al., 2002). Because only expression of wild-type HRSL3 was able to interfere with PP2A activity, we now addressed a potential role of the HRSL3-PR65α interaction in cell death. OVCAR-3 cells were transfected with wild-type HRSL3 and with the N-terminal deletion construct, then caspase cleavage and the DNA content were analyzed (Fig. 4A,B). Cleavage of caspase-3 was observed 48 hours after transfection with wild-type HRSL3 and could be completely inhibited in cells pretreated with the caspase inhibitor zVAD-fmk (Fig. 4A). The fraction of cells showing a subG1-characteristic DNA content was increased after transfection with HRSL3 by approximately 23%. The N-terminal deletion construct did not induce DNA fragmentation above background, as determined by the empty vector pcDNA3 (Fig. 4B). The pan-caspase inhibitor zVAD-fmk abrogated DNA fragmentation induced by HRSL3, confirming an involvement of caspases in this process (Fig. 4B, HREV1FL + zVAD). These observations indicated that inhibition of PP2A by the tumor suppressor HRSL3 might be the crucial event in the regulation of cell survival in ovarian carcinomas. To test whether inhibition of the phosphatase alone would induce apoptosis in ovarian carcinoma cells, we treated OVCAR-3 cells with 10 nM OA and measured the fraction of cells with a subG1 DNA content (Fig. 4B, right panel). In concert with published data showing a survival role of PP2A (Li et al., 2002; MacKeigan et al., 2005), after OA application more than 80% of the cells displayed a subG1 DNA content, which was reduced to 30% when the caspase-inhibitor zVAD-fmk was added (Fig. 4B).

  Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

HRSL3 suppresses the PP2A catalytic activity. (A) OVCAR-3 cells were transiently transfected with HREV1FL and HREV1-dNdC, transfection with pcDNA3 was used as a negative control, and treatment with 10 nM okadaic acid as a positive control. Forty-two hours after transfection cells were lysed and immunoprecipitated with an anti-PR65α antibody. Phosphatase activity of the immunoprecipitates was measured as the release of free phosphate (pmol/minute; y axis). Six independent experiments were performed, and included into a statistical analysis. Significance of the obtained results was assessed using an F-test. The difference between HREV1FL and dNdC, and HREV1FL and pcDNA3 values was considered as significant (P=0.02). (B) Immunoprecipitates used for the measurement of the phosphatase activity in the presence of HRSL3 and its interaction-deficient mutant were controlled by western blotting using HRSL3- and PR65-specific antibodies.

In order to prove a direct role of the HRSL3 interaction partner PR65α in preventing apoptosis in OVCAR-3 cells, we suppressed the PR65α subunit via RNAi and subsequently investigated caspase-3 cleavage (Fig. 4C, upper panel). As a control, we also performed a knock-down of PR65β, the efficiency of which was measured by RT-PCR (Fig. 4C, bottom panel). Cleaved caspase-3 was detected 48 hours after transfection of PR65α-specific siRNAs, whereas siRNA-mediated inhibition of the PR65β isoform induced a weaker activation of caspase-3. The caspase inhibitor zVAD-fmk prevented caspase cleavage during the siRNA experiments (Fig. 4C, upper panel), indicating that inhibition of PP2A, whether mediated through HRSL3 or via siRNA is sufficient to activate the caspase cascade in OVCAR-3 cells. Further measurement of the DNA content in siRNA-treated cells demonstrated that the PR65α-specific RNAi induced DNA fragmentation in approximately 49% of the cells, 29% were affected by inhibition of PR65β (Fig. 4D, upper panel), whereas a strong induction of DNA fragmentation (83%) was achieved after siRNA-dependent downregulation of both isoforms. Transfection with scrambled control siRNA induced DNA fragmentation in ∼24% of the cells (Fig. 4D, upper panel). Concomitant application of the pan-caspase inhibitor zVAD in each case strongly reduced the fraction of cells exhibiting a subG1-characteristic DNA content (Fig. 4D, middle panel). These results further demonstrate that both individual downregulation of PR65α and HRSL3 overexpression are sufficient to induce caspase-dependent apoptosis in ovarian carcinoma cells. Taking into account the observed decrease of the PR36 protein in the PR65 immunocomplex in the presence of HRSL3, we would speculate that HRSL3 might disrupt the PR65-PR36 protein complex thereby inhibiting PP2A activity and inducing apoptosis.

Interaction of HRSL3 with PR65α does not influence stability of PP2A

It has previously been demonstrated that ablation of either the regulatory or the catalytic subunits induces degradation of the whole PP2A enzyme (Li et al., 2002; Silverstein et al., 2002; Strack et al., 2004). Assuming that HRSL3 acts via a disruption of the PR65-PR36 interaction, the level of intracellular PR36 should be decreased in the presence of HRSL3. To prove this hypothesis, we analyzed the amounts of the PR36 and PR65 proteins in OVCAR-3 cells after PR65- and PR36-specific RNAi and after transfection with HRSL3. As shown in Fig. 5A, PR65α,β-specific RNAi leads to a reduction in the PR36 protein level. In the control experiment, PR36-specific RNAi leads to a decrease in the relative amount of the PR65 protein. By contrast, HRSL3 has no effect on the protein level of either the catalytic or the regulatory subunit of PP2A (Fig. 5B), albeit we demonstrated that the amount of PR36 in the PR65 immunocomplex decreased in the presence of HRSL3. This suggests that HRSL3 might change the composition of the PP2A protein complex, yet does not suppress PP2A activity by inducing its degradation.

Atypical protein kinase C (PKC) is potentially involved in HRSL3 signaling in ovarian carcinoma cells

To decipher the apoptotic pathway induced by HRSL3-mediated suppression of PP2A in OVCAR-3 cells in more details, we analyzed the amounts and phosphorylation of anti-apoptotic Bcl and pro-apoptotic Bad, which are known to be PP2A targets (Garcia et al., 2003; Grethe and Porn-Ares, 2005). Western blot analysis revealed that neither HRSL3, nor administration of OA can influence phosphorylation and relative levels of these proteins in OVCAR-3 cells (Fig. 6A).

To further define the caspases involved in apoptosis induction, we performed western blot analysis of cleaved caspases after HRSL3 transfection and OA treatment. We did not detect cleavage of the initiator caspase-8 and caspase-10 after HRSL3 transfection, although caspase-8 was slightly activated by OA (data not shown). By contrast, caspase-9 was cleaved after transfection with the HRSL3 expression vector and after OA treatment (Fig. 6A, left panel). Activation of caspase-9 resulted in the cleavage of its effector caspases-7 and 3. Transfection with the empty vector or treatment with the drug vehicle, DMSO, did not affect any of the caspases examined (Fig. 6A, left panel). This result suggests that HRSL3 induces apoptosis via a caspase-9-dependent, but caspase-8- and 10-independent pathway.

  Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Both HRSL3-dependent PP2A inhibition and siRNA-mediated suppression of PP2A regulatory subunit A, induce apoptosis via a caspase-dependent pathway. (A) Cleaved caspase-3 was detected by immunoblotting using appropriate antibodies. Total amount of caspase-3 was used as a loading control. In all experiments HRSL3 expression was controlled by immunoblotting with the HRSL3 specific antibody. Application of the caspase inhibitor zVAD-fmk leads to the abrogation of caspase cleavage. (B) OVCAR-3 cells were electroporated with the HREV1FL, dNdC expression plasmids. Twelve hours after transfection, cells were treated with 40 μM caspase inhibitor zVAD-fmk, if indicated. Alternatively, cells were treated with OA (10 nM), OA with zVAD-fmk (40 μM), or vehicle DMSO only. Seventy-two hours after transfection or treatment, nuclear DNA content was measured by flow cytometry. The relative number of cells displaying an apoptotic, sub-G1 content, is given between the marker bars. DNA histograms are representative of two independent experiments. (C) OVCAR-3 cells were transfected with PR65α- and PR65β-specific siRNAs individually, together, and treated with caspase inhibitor zVAD-fmk (40 μM), if indicated. The level of the PR65α protein and cleavage of caspase-3 were controlled 48 hours after transfection (upper panel). Efficiency of the PR65-specific RNA interference was verified using RT-PCR 12 and 24 hours after transfection. As a control, GAPDH mRNA was amplified (lower panel). (D) OVCAR-3 cells were transfected with PR65α- and PR65β-specific siRNAs individually or in combination. Apoptosis was analyzed at the single-cell level by measuring the nuclear DNA content. The relative number of cells displaying a sub-G1 DNA content is given between the marker bars. DNA histograms are representative of three independent experiments.

Next, we analyzed activation and phosphorylation of RAF-MAPK, PI 3-kinase, and Wnt pathways through HRSL3 and OA in OVCAR-3 cells, because PP2A is known to be involved in the regulation of these signaling cascades (Alessi et al., 1995; Chen et al., 2005; Gao et al., 2002; Gotz et al., 2000; Ikeda et al., 2000; Li et al., 2003; Li et al., 2001; Ratcliffe et al., 2000; Ricciarelli and Azzi, 1998; Seeling et al., 1999; Sim and Scott, 1999; Yamamoto et al., 2001; Yang et al., 2003; Zhou et al., 2002; Zuluaga et al., 2007). Treatment with OA led to the activation of ERK1,2, AKT (PKB), and β-catenin (Fig. 6A, right panel), whereas neither RAF no PDK seemed to be regulated by OA-dependent phosphatases. In contrast to OA, HRSL3 did not affect phosphorylation of ERK and AKT (PKB), which is consistent with our previous observation that HRSL3 only partially inhibits PP2A catalytic activity (Fig. 3A).

We asked if an HRSL3-independent partial inhibition of PP2A might be sufficient to induce apoptosis in ovarian carcinoma cells. Since the IC50 of OA varies depending on the substrate employed and on the amount of the enzyme, both under cell-free conditions and in cellular systems (Boudreau and Hoskin, 2005; Favre et al., 1997; McCluskey et al., 2002; Schonthal, 1998), we empirically defined the OA concentrations required for apoptosis in OVCAR-3 cells. 0.5 nM OA was sufficient to induce caspase cleavage, with an efficiency similar to that of HRSL3 expression (Fig. 6B, right panel), confirming that partial inhibition of PP2A is sufficient to induce the apoptotic cascade in ovarian carcinoma cells. In addition, treatment of the cells with 1 nM OA did not significantly influence phosphorylation of ERK1/2 and MEK1/2 (Fig. 6B, left panel), suggesting that at this concentration, the cells maintain a pool of active PP2A which dephosphorylates the kinases analyzed.

Most interestingly, a clear increase in the phosphorylation of the atypical PKCζ was detected 48 hours post-transfection with full-length HRSL3, but not with the empty vector or with the vehicle (Fig. 6C). Thus PKCζ is a PP2A target, whose phosphorylation is regulated by both OA and HRSL3, suggesting a potential implication of PKC in HRSL3-dependent signaling. To prove whether PKCζ is involved in HRSL3-dependent apoptosis, we utilized a myristilated pseudosubstrate peptide, which has been shown to specifically inhibit PKCζ (Standaert et al., 1999). Cells, pre-treated with different concentrations of the peptide, were transfected with the HRSL3 expression plasmid, and then caspase-3 cleavage was analyzed. As shown in Fig. 6D, PKCζ inactivation abrogated HRSL3-induced apoptosis to the background level, suggesting that PKCζ activity might be critical for the pro-apoptotic HRSL3 function in ovarian cancer cells.

Discussion

Our previous work has demonstrated a tumor suppressing role of the HRSL3 protein in ovarian carcinomas (Sers et al., 2002). In an attempt to define the underlying mechanism, we could now show that in OVCAR-3 ovarian cancer cells HRSL3 directly interacts with the PP2A regulatory subunit Aα. HRSL3 ablates the catalytic activity of the PP2A holoenzyme, which finally results in a caspase-9-dependent cell death. Failure of an HRSL3 interaction-deficient mutant to promote apoptosis let us to suggest that formation of the HRSL3 and PR65α protein complex is a key event in HRSL3-mediated apoptosis. A preliminary analysis of several PP2A target molecules revealed that the HRSL3-PP2A interaction specifically abrogates the ability of PP2A to dephosphorylate atypical PKCζ, the activity of which seems to be critical for HRSL3 function. This suggests an involvement of PKCζ in the regulation of cell survival in ovarian cancer.

  Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

HRSL3 does not influence the stability of PP2A. (A,B) OVCAR-3 cells were transfected with PR36α-, PR36β-, PR65α- or PR65β-specific siRNAs, or with the HRSL3-expressing plasmid (HREV1FL). Relative amounts of the proteins were controlled 24 and 48 hours after transfection using western blotting against PR65 or PR36 proteins.

Tumor suppressing function of the H-REV107-like proteins

HRSL3 belongs to the H-REV107-like family of tumor suppressors (Hughes and Stanway, 2000). Although the biochemical properties of these proteins are still elusive, there is ample evidence that H-REV107-like proteins are involved in the regulation of cell proliferation, differentiation and apoptosis. HRSL3 was found to suppress growth of rodent and human carcinoma cells in vitro and in vivo and is downregulated in breast, ovary and kidney carcinomas (Husmann et al., 1998; Sers et al., 1997; Sers et al., 2002). The RARRES3 protein is suppressed in many human tumors such as ovarian (Lotz et al., 2005), gastric (Huang et al., 2000), colorectal (Huang et al., 2000; Shyu et al., 2003) and head and neck tumors (Higuchi et al., 2003), in psoriatic lesions and in basal cell carcinomas (Duvic et al., 2000; Duvic et al., 2003). It can activate type I transglutaminase by an unknown mechanism thereby inducing terminal differentiation (Sturniolo et al., 2003). In cervical cancer cells, RARRES3 suppresses the activation of RAS proteins (Tsai et al., 2006) and negatively regulates the activity of Jun and p38 MAPKs (Huang et al., 2002). Interestingly, both HRASL3 and RARRES3 were identified as downstream targets of IFNγ (Sers et al., 2002) and are suggested to regulate ovarian carcinoma cell growth by a concerted action (Lotz et al., 2005). However, whereas RARRES3 has been identified as a suppressor of RAS and MAPK activation (Tsai et al., 2006), HRSL3 negatively affects PP2A, thereby specifically targeting PKC, independent of MAPK and PI 3-kinase activation. This indicates that the concerted loss of HRSL3 and RARRES3 from advanced ovarian carcinomas (Lotz et al., 2005; Sers et al., 2002) might allow a sustained activation of several distinct branches of the intracellular oncogenic signaling network.

  Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

Expression of the full-length HRSL3 protein (HREVFL) was controlled by incubation with the HRSL3-specific antibody (H-REV107-1). HRSL3 regulates PKCζ, which is required for HRSL3-induced apoptosis. (A) Phosphorylation of the indicated signaling proteins was analyzed by immunoblotting using antibodies specific for their phosphorylated forms. Immunoblotting against total amount of the proteins was used as a loading control. Expression of the HRSL3 protein, full-length and truncated form, was controlled by incubation with the HRSL3-specific antibody. (B) OVCAR-3 cells were either treated with 1, 10 and 20 nM of OA and with an inhibitor of the PI 3-kinase (LY294002; LY), or transfected with HRSL3. To determine the concentration of OA that inhibits PP2A activity only partially, phosphorylation of MEK1/2 and ERK1/2 kinases was examined using the corresponding antibodies (left panel). Then caspase-9 cleavage was analyzed (right panel). Actin- and caspase-9-specific antibodies were used as loading controls. (C) Phosphorylation of PKCζ is induced by treatment with both OA and HRSL3. Cells were treated with 10 nM OA, with DMSO or transfected with HRSL3 or the empty vector. Phospho-PKCζ or total PKCζ were measured by western blotting using the appropriate antibodies. (D) Twelve hours prior to transfection, OVCAR-3 cells were treated with the myristoylated PKCζ-specific pseudosubstrate. Then the cells were transfected with HRASL3 expression plasmid and caspase cleavage was analyzed 48 hours after transfection.

In a recent publication, Anantharaman and Aravind proposed that H-REV107-like proteins belong to the NlpC/P60 protein superfamily, consisting largely of prokaryotic members, most of which act as peptidases. Lecithin retinol acyltransferase (LRAT), an enzyme responsible for the esterification of retinol to retinyl ester (Ruiz et al., 1999), was suggested to be the founder of the eukaryotic members of this superfamily (Anantharaman and Aravind, 2003). Similar to H-REV107-like proteins, LRAT expression is reduced in several human tumors and in corresponding cell lines (Andreola et al., 2000; Boorjian et al., 2004; Guo et al., 2000; Guo et al., 2001; Guo et al., 2002; Guo and Gudas, 1998; Jurukovski and Simon, 1999; Simmons et al., 2002; Zhan et al., 2003). In contrast to the H-REV107 proteins and most of the prokaryotic related proteins, LRAT has a defined biochemical function and the active motifs within the protein have been characterized (Mondal et al., 2000; Mondal et al., 2001; Xue and Rando, 2004). Mutation of the critical residue Cys112, which is conserved between LRAT and HRSL3, had no influence on binding and suppression of PP2A by HRSL3. This suggests that the HRSL3 protein is not likely to exert an acyl-transferase function, and also leaves open to what extend the conservation of distinct domains between these proteins reflects a functional similarity. Most importantly, LRAT has been found recently also to harbor a palmitoylation function, indicating that this protein family might have a number of different biochemical properties (Xue et al., 2006). Since PP2A activity is controlled by post-translational modifications such as phosphorylation and methylation, it is an attractive hypothesis that HRSL3 might exert a protein modifying function (Bryant et al., 1999; Janssens and Goris, 2001; Turowski et al., 1995; Wu et al., 2000), however, currently, no data supporting a possible enzymatic activity of H-REV107 proteins are available.

A role for HRSL3 in controlling the PR65 protein complex

We detected the ectopically expressed HRSL3 protein in a complex with the endogenous PR65α protein in OVCAR-3 cells and could prove in vitro interaction between HRSL3 and PP2A. More specifically, we showed that the amount of the catalytic subunit PR36α recovered from the PR65 protein complex is considerably reduced in the presence of HRSL3. Additionally, PR36 and HRSL3, despite their predominant cytoplasmic distribution, were not colocalized within the cell. This suggests, but does not prove, that HRSL3 might somehow substitute for PR36 in a distinct pool of PP2A holoenzymes. Additionally, the absence of PR36 protein degradation following HRSL3 overexpression suggests that upon depletion of PR36 from the PR65 protein complex, PR36 binds to other interaction partners such as alpha-4 or cyclin G2 known to directly interact with PR36 thereby protecting it from degradation (Bennin et al., 2002; Chen et al., 1998; Chung et al., 1999; Prickett and Brautigan, 2004).

Considering the complexity of multiple PP2A holoenzymes coexisting within a cell (Janssens and Goris, 2001; Sontag, 2001), HRSL3 is likely to interact with and to inhibit the activity of only a specific subset of PP2A holoenzymes. It is well known that different PP2A holoenzymes simultaneously interact with distinct targets and regulatory proteins. The composition of the PP2A heterotrimers determines their localization and their functional specificity. For instance, the PP2AB56C phosphatase interacts with the β-catenin degradation complex and inhibits Wnt-signaling in Drosophila (Li et al., 2001), whereas PP2AB56ϵC positively regulates the Wnt pathway during early Xenopus embryogenesis (Yang et al., 2003). When we addressed the activation status of several distinct signaling cascades downstream of the HRSL3 protein, we could show that inhibition of PP2A activity via HRSL3 activates apoptosis in a caspase-9-dependent manner. PP2A inhibition via OA or PP2A-specific RNAi also led to cell death, pointing to a role of the phosphatase in survival of ovarian carcinomas. This observation is supported by many reports showing that PP2A acts as one of the key regulators of cell proliferation and malignant transformation in other cell systems (Abraham et al., 2000; Chen et al., 2004; Chen et al., 2005; Jaumot and Hancock, 2001; Li et al., 2002).

Analysis of the PP2A substrates revealed PKCζ, shown earlier to be activated by OA in rat adipocytes, (Standaert et al., 1999), as a potential HRSL3 target in ovarian carcinoma cells. Importantly, treatment of cells with the PKCζ-specific pseudosubstrate abolished HRSL3-dependent caspase cleavage, supporting the possibility that PKCζ activity might be of functional relevance for apoptosis induction. The first evidence of a cross-talk between PP2A and PKCζ was provided by Sontag et al., who demonstrated that during an SV40 infection, inhibition of PP2A is essential for the activation of PKCζ and NF-κB signaling pathways in monkey kidney CV-1 cells (Sontag et al., 1997). Furthermore, PP2A was shown to regulate atypical PKCs during tight junction formation in epithelial cells (Nunbhakdi-Craig et al., 2002). Recently, the atypical PKCζ and the novel PKCθ were identified to be upregulated in ovarian carcinoma; PKCζ was suggested as a potential biomarker for aggressive disease (Zhang et al., 2006). Our current data suggest an additional role for PKCζ-dependent signal transduction in the progression of ovarian carcinoma and implicate HRSL3 as a signaling regulatory molecule functionally involved in the complex regulatory network mediating growth and survival of ovarian cancer.

Materials and Methods

Yeast two-hybrid analysis

A LexA-based yeast two-hybrid screening of a human kidney cDNA library was performed according to the recommendations of the manufacturer (BD Biosciences, Clontech, CA). The EGY48 yeast strain (OriGene Technologies, Inc., MD) was consecutively transformed with a p8op-lacZ reporter plasmid (BD Biosciences, Clontech, CA), a pEG202 plasmid (Pharmacia Biotech Inc., CA) carrying the HRSL3 cDNA, and a pJG4-5 plasmid containing a library insert (OriGene Technologies Inc, MD). Positive clones were selected on the SD, Gal, Raf, –His, –Leu, –Trp, –Ura, X-gal induction medium. Plasmid DNA was isolated from all positive clones, transferred into the KC8 bacterial strain and sequenced.

Expression constructs and siRNAs

HREV1FL was described previously (Hajnal et al., 1994). HREV1dCHA was generated by PCR amplification. It harbors the first 406 bp of the HRSL3 coding region, and 28 additional basepairs encoding an artificial HA-epitope (YPYDADYA). HREV1-AWAIdCHA (His23→Ala23) and HREV1-NSEdCHA (Cys112→Ser112) were generated using the QuikChange Mutagenesis Kit (Stratagene, CA). The dNdCHA construct harbors a deletion of 30 bp at the 5′-end of the HRSL3 cDNA coding region. An expression vector containing human PR65α fused to a V5 epitope (PR65αV5) was purchased from GeneStorm Collection (Invitrogen, CA). The pcDNA3 (Invitrogen, CA) plasmid was used as a control. PR65α,β-, PR36α,β-specific siRNAs and control duplexes were purchased from the Validated Stealth™ RNAi Collection (Invitrogen, CA).

RT-PCR

RT-PCR reactions were carried out using Access RT-PCR System according to the recommendations of the supplier (Promega, WI). Primers used for the amplification of 211 bp of the PPP2R1A gene (NM_014225) were 5′-CGAACTCCGCAATGAGGACG-3′ (fw), and 5′-AGGCAGTGCACGTACTCTGG-3′ (rv), and for the amplification of 260 bp the PPP2R1B gene (NM 002716) were 5′-CGACGAGCTCCGCAATGAAG-3′ (fw) and 5′-CAACAGTCTCTTCCACAGTTGCC-3′ (rv). As a control a G3PDH-specific fragment was amplified using primers 5′-GAACGGGAAGCTTGTCATTCA-3′ (fw) and 5′-GTAGCCAAATTCGTTGTCATAC-3′ (rv).

Cell culture and transfection procedures and treatment

Human epithelial ovarian carcinoma cell lines SKOV-3, OVCAR-3, CAOV-3, A27/80, the teratocarcinoma cell line PA1, the cervix adenocarcinoma cell line HeLa, the epidermoid carcinoma cell line A431, the African green monkey kidney fibroblasts COS-7, and the H-Ras transformed rat fibroblasts FE-8 (Griegel et al., 1986) were maintained in Dulbecco's modified Eagle's medium (DMEM; BioWhittaker, MD) supplemented with 10% fetal calf serum and 2 mM glutamine. DNA transfection was performed using Fugene reagent (Roche Diagnostics GmbH, Mannheim, Germany); siRNA transfection was done using Lipofectamine 2000 (Invitrogen, CA). For the analysis of the endogenous PR65α protein complex and DNA fragmentation, HRSL3 expression plasmids (HREV1FL and dNdC) were transfected into OVCAR-3 cells using an Amaxa device for electroporation (Amaxa Biosystems, Cologne, Germany) in solution R and the T16-program. For cell treatment, okadaic acid (Upstate, CA) and a myristoylated PKCzeta-specific pseudosubstrate inhibitor Myr-SIYRRGARRWRKL-OH, synthesized at the Peptide-synthesis Service Facility (Institute of Biochemistry, Charité, Berlin, Germany) were used.

GST pull-down assay

GST-HREVdC fusion protein and GST control proteins were produced in B21 strain of E. coli and purified using gluthatione-Sepharose. Subsequently, OVCAR-3 cells were lysed in NP40-lysis buffer [50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% Nonidet P40, 10% glycerol, EDTA-free protease inhibitor complete tabs (Roche Diagnostics GmbH, Mannheim, Germany)]. Clarified protein extracts were incubated for 1 hour with GST-HREVdC and GST alone coupled to glutathione-Sepharose, washed four times with lysis buffer and analyzed using SDS-PAGE for presence of the PR65 protein.

Immunoprecipitation and western blot analysis

Transfected COS-7 cells were lysed in NP40-lysis buffer. Clarified lysates were used for immunoprecipitation with either anti-V5 or anti-HA antibody (Roche Diagnostics GmbH, Mannheim, Germany). Precipitated protein complexes were washed three times with lysis buffer and analyzed by SDS-PAGE. To immunoprecipitate the endogenous PR65 and PR36 protein complex, OVCAR-3 cells were lysed in Triton lysis buffer (50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% Triton X-100, 0.5 mM PMSF, EDTA-free protease inhibitor complete tabs); clarified protein lysates were precipitated either with a PR65α-specific antibody (Covance Research Products Inc, CA) or with PR36αβ-specific antibody (Upstate Biotechnology, NY). Precipitates were washed and subjected to SDS-PAGE. Alternatively, for the isolation of the PR65α immunocomplex (Fig. 3A), the ProFound Mammalian Co-immunoprecipitation Kit was used (Pierce Biotechnology Inc., IL). The experimental procedure was performed according to the recommendations of the supplier.

To detect cleaved caspases, cells were harvested in cold 1× PBS, and lysed in Chaps buffer (50 mM Pipes-HCl pH 6.5, 2 mM EDTA, 0.1% Chaps, 20 μg/ml leupeptin, 10 μg/ml pepstatin A, μg/ml aprotinin, 5 mM DTT). To disrupt the cells, three freeze-thaw cycles were used.

For western blot analyses, proteins were separated by SDS-PAGE, and transferred to a PVDF membrane (Amersham Pharmacia, Buckinghamshire, UK). Membranes were blocked and incubated with the following primary antibodies: anti-V5 (Invitrogen, CA), anti-HA (Sigma, MS), anti-PR65α (6F9; Covance Research Products Inc, CA), anti-PR65 and anti-PR36α (BD Biosciences, Heidelberg, Germany), anti-pan-actin (Chemicon, CA) and anti-HRSL3 (Sers et al., 2002). All other antibodies were purchased from Cell Signaling Technology Inc. (MA). Then the membranes were incubated with a peroxidase-conjugated goat anti-rabbit, goat anti-rat or goat anti-mouse antisera (Dianova, Hamburg, Germany) and developed using the ECL System (Amersham Pharmacia, Buckinghamshire, UK).

Immunofluorescence analysis and confocal microscopy

For immunofluorescence analysis cells were grown on glass coverslips. After fixation with 3% paraformaldehyde, cells were permeabilized using 0.2% Triton X-100 for 1 min, then washed with PBS and incubated with the primary antibodies against HRSL3, PR65 or PR36 (BD Biosciences, Heidelberg, Germany), in 1% BSA-PBS, washed and incubated with secondary antibodies, anti-rabbit Alexa Fluor 594, Alexa Fluor 546, anti-mouse Alexa Fluor 488 (Molecular Probes, OR), or anti-rat FITC (The Jackson Laboratory, MA). For nuclear staining, cells were incubated with diamidinophenylindole (DAPI). Colocalization of HREV1FL with PR65 and PR36 was analyzed using a Leica confocal microscope TCS SL (×63 oil objective, 2× zoom).

Phosphatase assay

Phosphatase activity was measured using the Ser/Thr Phosphatase Assay Kit (Upstate Biotechnology, NY). Cells were lysed in lysis buffer (50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% Nonidet P40, 10% glycerol, EDTA-free protease inhibitor complete tabs) and equal amount of clarified lysates were precipitated with an anti-PR65α antibody and protein G-agarose (Roche, Mannheim, Germany). For each reaction, 250 μM phosphopeptide K-R-pT-I-R-R was used as a substrate. The amount of free phosphate was determined in comparison with the standard curve and calculated as pmol/minute. The results of six independent experiments were used for statistical analysis using F-tests (MS Office Excel).

Measurement of apoptotic cell death

Apoptosis was determined at the single-cell level by measuring the DNA content of individual cells. The protocol was adapted from that of Hemmati et al. (Hemmati et al., 2002). Briefly, OVCAR-3 cells were trypsinized, collected and fixed in 2% paraformaldehyde. Cells were incubated in 70% ethanol and resuspended in PBS containing 40 mg/ml DNase-free RNase A (Roche Molecular Biochemicals, Mannheim, Germany). After incubation for 30 minutes cells were pelleted and resuspended in PBS containing 50 mg/ml propidium iodide (Sigma, Deisenhofen, Germany). Cellular DNA content was measured with a logarithmic amplification in the FL-3 channel of a FACScan flow cytometer (BD, Heidelberg, Germany) equipped with the CELL-Quest software. Data are expressed in percent hypoploidy (the percentage of cells with a sub-G1 DNA content), which reflects the percentage of apoptotic cells with fragmented genomic DNA.

Acknowledgments

We are grateful for the technical support from Cornelia Gieseler, Berit Lenz, Danny Bashin, Jana Keil, Kristin Lucht, Jenny Eichhorst, and Anja and Antje Richter. The help of Matthias Truss, Wiljan Hendriks and Edwin Cuppen with protein interaction, Peter Daniel with DNA fragmentation studies, and Christine Rueckert and Burkhard Wissner with confocal microscopy is greatly acknowledged. Our project was supported by the Deutsche Krebshilfe (grants 10-1273 SE1, 10-1803 SE2 to C.S.) and SFB 506 to R.S. and C.S.

Footnotes

  • Supplementary material available online at http://jcs.biologists.org/cgi/content/full/120/8/1393/DC1

  • ↵* Present address: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany

  • Accepted January 29, 2007.
  • © The Company of Biologists Limited 2007

References

  1. ↵
    Abraham, D., Podar, K., Pacher, M., Kubicek, M., Welzel, N., Hemmings, B. A., Dilworth, S. M., Mischak, H., Kolch, W. and Baccarini, M. (2000). Raf-1-associated protein phosphatase 2A as a positive regulator of kinase activation. J. Biol. Chem. 275, 22300-22304.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    Alessi, D. R., Gomez, N., Moorhead, G., Lewis, T., Keyse, S. M. and Cohen, P. (1995). Inactivation of p42 MAP kinase by protein phosphatase 2A and a protein tyrosine phosphatase, but not CL100, in various cell lines. Curr. Biol. 5, 283-295.
    OpenUrlCrossRefPubMedWeb of Science
  3. ↵
    Anantharaman, V. and Aravind, L. (2003). Evolutionary history, structural features and biochemical diversity of the NlpC/P60 superfamily of enzymes. Genome Biol. 4, R11.
    OpenUrlCrossRefPubMed
  4. ↵
    Andreola, F., Giandomenico, V., Spero, R. and De Luca, L. M. (2000). Expression of a smaller lecithin:retinol acyl transferase transcript and reduced retinol esterification in MCF-7 cells. Biochem. Biophys. Res. Commun. 279, 920-924.
    OpenUrlCrossRefPubMedWeb of Science
  5. ↵
    Bastians, H. and Ponstingl, H. (1996). The novel human protein serine/threonine phosphatase 6 is a functional homologue of budding yeast Sit4p and fission yeast ppe1, which are involved in cell cycle regulation. J. Cell Sci. 109, 2865-2874.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Bennin, D. A., Don, A. S., Brake, T., McKenzie, J. L., Rosenbaum, H., Ortiz, L., DePaoli-Roach, A. A. and Horne, M. C. (2002). Cyclin G2 associates with protein phosphatase 2A catalytic and regulatory B' subunits in active complexes and induces nuclear aberrations and a G1/S phase cell cycle arrest. J. Biol. Chem. 277, 27449-27467.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Boorjian, S., Tickoo, S. K., Mongan, N. P., Yu, H., Bok, D., Rando, R. R., Nanus, D. M., Scherr, D. S. and Gudas, L. J. (2004). Reduced lecithin: retinol acyltransferase expression correlates with increased pathologic tumor stage in bladder cancer. Clin. Cancer Res. 10, 3429-3437.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Boudreau, R. T. and Hoskin, D. W. (2005). The use of okadaic acid to elucidate the intracellular role(s) of protein phosphatase 2A: lessons from the mast cell model system. Int. Immunopharmacol. 5, 1507-1518.
    OpenUrlCrossRefPubMed
  9. ↵
    Brewis, N. D., Street, A. J., Prescott, A. R. and Cohen, P. T. (1993). PPX, a novel protein serine/threonine phosphatase localized to centrosomes. EMBO J. 12, 987-996.
    OpenUrlPubMedWeb of Science
  10. ↵
    Bryant, J. C., Westphal, R. S. and Wadzinski, B. E. (1999). Methylated C-terminal leucine residue of PP2A catalytic subunit is important for binding of regulatory Balpha subunit. Biochem. J. 339, 241-246.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Chen, J., Peterson, R. T. and Schreiber, S. L. (1998). Alpha 4 associates with protein phosphatases 2A, 4, and 6. Biochem. Biophys. Res. Commun. 247, 827-832.
    OpenUrlCrossRefPubMedWeb of Science
  12. ↵
    Chen, W., Possemato, R., Campbell, K. T., Plattner, C. A., Pallas, D. C. and Hahn, W. C. (2004). Identification of specific PP2A complexes involved in human cell transformation. Cancer Cell 5, 127-136.
    OpenUrlCrossRefPubMedWeb of Science
  13. ↵
    Chen, W., Arroyo, J. D., Timmons, J. C., Possemato, R. and Hahn, W. C. (2005). Cancer-associated PP2A Aalpha subunits induce functional haploinsufficiency and tumorigenicity. Cancer Res. 65, 8183-8192.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Chung, H., Nairn, A. C., Murata, K. and Brautigan, D. L. (1999). Mutation of Tyr307 and Leu309 in the protein phosphatase 2A catalytic subunit favors association with the alpha 4 subunit which promotes dephosphorylation of elongation factor-2. Biochemistry 38, 10371-10376.
    OpenUrlCrossRefPubMed
  15. ↵
    Cohen, P. T., Chen, M. X. and Armstrong, C. G. (1996). Novel protein phosphatases that may participate in cell signaling. Adv. Pharmacol. 36, 67-89.
    OpenUrlCrossRefPubMed
  16. ↵
    Deucher, A., Nagpal, S., Chandraratna, R. A. S., Di Sepio, D., Robinson, N. A., Dashti, S. R. and Eckert, R. L. (2000). The carboxy-terminal hydrophobic domain of TIG3, a class II tumor suppressor protein, is required for appropriate cellular localization and optimal biological activity. Int. J. Oncol. 17, 1195-1203.
    OpenUrlPubMedWeb of Science
  17. ↵
    DiSepio, D., Ghosn, C., Eckert, R. L., Deucher, A., Robinson, N., Duvic, M., Chandraratna, R. A. S. and Nagpal, S. (1998). Identification and characterization of a retinoid-induced class II tumor suppressor growth regulatory gene. Proc. Natl. Acad. Sci. USA 95, 14811-14815.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Duvic, M., Helekar, B., Schulz, C., Cho, M., DiSepio, D., Hager, C., Dimao, D., Hazarika, P., Jackson, B., Breuer-McHam, J. et al. (2000). Expression of a retinoid-inducible tumor suppressor, Tazarotene-inducible gene-3, is decreased in psoriasis and skin cancer. Clin. Cancer Res. 6, 3249-3259.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Duvic, M., Ni, X., Talpur, R., Herne, K., Schulz, C., Sui, D., Ward, S., Joseph, A. and Hazarika, P. (2003). Tazarotene-induced gene 3 is suppressed in basal cell carcinomas and reversed in vivo by tazarotene application. J. Invest. Dermatol. 121, 902-909.
    OpenUrlCrossRefPubMed
  20. ↵
    Favre, B., Turowski, P. and Hemmings, B. A. (1997). Differential inhibition and posttranslational modification of protein phosphatase 1 and 2A in MCF7 cells treated with calyculin-A, okadaic acid, and tautomycin. J. Biol. Chem. 272, 13856-13863.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Gao, Z. H., Seeling, J. M., Hill, V., Yochum, A. and Virshup, D. M. (2002). Casein kinase I phosphorylates and destabilizes the beta-catenin degradation complex. Proc. Natl. Acad. Sci. USA 99, 1182-1187.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Garcia, A., Cayla, X., Guergnon, J., Dessauge, F., Hospital, V., Rebollo, M. P., Fleischer, A. and Rebollo, A. (2003). Serine/threonine protein phosphatases PP1 and PP2A are key players in apoptosis. Biochimie 85, 721-726.
    OpenUrlCrossRefPubMed
  23. ↵
    Gotz, J., Probst, A., Mistl, C., Nitsch, R. M. and Ehler, E. (2000). Distinct role of protein phosphatase 2A subunit Calpha in the regulation of E-cadherin and beta-catenin during development. Mech. Dev. 93, 83-93.
    OpenUrlCrossRefPubMedWeb of Science
  24. ↵
    Grethe, S. and Porn-Ares, M. I. (2005). p38 MAPK regulates phosphorylation of Bad via PP2A-dependent suppression of the MEK1/2-ERK1/2 survival pathway in TNF-alpha induced endothelial apoptosis. Cell. Signal. 18, 531-540.
    OpenUrlCrossRefPubMed
  25. ↵
    Griegel, S., Traub, O., Willecke, K. and Schäfer, R. (1986). Suppression and re-expression of transformed phenotype in hybrids of HA-ras-1-transformed rat-1 cells and early-passage rat embryonic fibroblasts. Int. J. Cancer 5, 697-705.
    OpenUrl
  26. ↵
    Guo, X. and Gudas, L. J. (1998). Metabolism of all-trans-retinol in normal human cell strains and squamous cell carcinoma (SCC) lines from the oral cavity and skin: reduced esterification of retinol in SCC lines. Cancer Res. 58, 166-176.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Guo, X., Ruiz, A., Rando, R. R., Bok, D. and Gudas, L. J. (2000). Esterification of all-trans-retinol in normal human epithelial cell strains and carcinoma lines from oral cavity, skin and breast: reduced expression of lecithin:retinol acyltransferase in carcinoma lines. Carcinogenesis 21, 1925-1933.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Guo, X., Nanus, D. M., Ruiz, A., Rando, R. R., Bok, D. and Gudas, L. J. (2001). Reduced levels of retinyl esters and vitamin A in human renal cancers. Cancer Res. 61, 2774-2781.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Guo, X., Knudsen, B. S., Peehl, D. M., Ruiz, A., Bok, D., Rando, R. R., Rhim, J. S., Nanus, D. M. and Gudas, L. J. (2002). Retinol metabolism and lecithin:retinol acyltransferase levels are reduced in cultured human prostate cancer cells and tissue specimens. Cancer Res. 62, 1654-1661.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Hajnal, A., Klemenz, R. and Schäfer, R. (1994). Subtraction cloning of H-Rev107, a gene specifically expressed in H-Ras resistant fibroblasts. Oncogene 9, 479-490.
    OpenUrlPubMedWeb of Science
  31. ↵
    Hemmati, P. G., Gillissen, B., von Haefen, C., Wendt, J., Starck, L., Guner, D., Dorken, B. and Daniel, P. T. (2002). Adenovirus-mediated overexpression of p14(ARF) induces p53 and Bax-independent apoptosis. Oncogene 21, 3149-3161.
    OpenUrlCrossRefPubMedWeb of Science
  32. ↵
    Higuchi, E., Chandraratna, R. A., Hong, W. K. and Lotan, R. (2003). Induction of TIG3, a putative class II tumor suppressor gene, by retinoic acid in head and neck and lung carcinoma cells and its association with suppression of the transformed phenotype. Oncogene 22, 4627-4635.
    OpenUrlCrossRefPubMed
  33. ↵
    Holmes, C. F., Luu, H. A., Carrier, F. and Schmitz, F. J. (1990). Inhibition of protein phosphatases-1 and -2A with acanthifolicin. Comparison with diarrhetic shellfish toxins and identification of a region on okadaic acid important for phosphatase inhibition. FEBS Lett. 270, 216-218.
    OpenUrlCrossRefPubMedWeb of Science
  34. ↵
    Huang, S. L., Shyu, R. Y., Yeh, M. Y. and Jiang, S. Y. (2000). Cloning and characterization of a novel retinoid-inducible gene 1 (RIG1) deriving from human gastric cancer cells. Mol. Cell. Endocrinol. 159, 15-24.
    OpenUrlCrossRefPubMed
  35. ↵
    Huang, S. L., Shyu, R. Y., Yeh, M. Y. and Jiang, S. Y. (2002). The retinoid-inducible gene I: Effect on apoptosis and mitogen-activated kinase signal pathways. Anticancer Res. 22, 799-804.
    OpenUrlPubMed
  36. ↵
    Hughes, P. J. and Stanway, G. (2000). The 2A proteins of three diverse picornaviruses are related to each other and to the H-rev107 family of proteins involved in the control of cell proliferation. J. Gen. Virol. 81, 201-207.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Husmann, K., Sers, C., Fietze, E., Mincheva, A., Lichter, P. and Schäfer, R. (1998). Transcriptional and translational downregulation of H-REV107, a class II tumour suppressor gene located on human chromosome 11q11-12. Oncogene 17, 1305-1312.
    OpenUrlCrossRefPubMedWeb of Science
  38. ↵
    Ikeda, S., Kishida, M., Matsuura, Y., Usui, H. and Kikuchi, A. (2000). GSK-3beta-dependent phosphorylation of adenomatous polyposis coli gene product can be modulated by beta-catenin and protein phosphatase 2A complexed with Axin. Oncogene 19, 537-545.
    OpenUrlCrossRefPubMedWeb of Science
  39. ↵
    Janssens, V. and Goris, J. (2001). Protein phosphatase 2A: a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling. Biochem. J. 353, 417-439.
    OpenUrlCrossRefPubMedWeb of Science
  40. ↵
    Janssens, V., Goris, J. and Van Hoof, C. (2005). PP2A: the expected tumor suppressor. Curr. Opin. Genet. Dev. 15, 34-41.
    OpenUrlCrossRefPubMedWeb of Science
  41. ↵
    Jaumot, M. and Hancock, J. F. (2001). Protein phosphatases 1 and 2A promote Raf-1 activation by regulating 14-3-3 interactions. Oncogene 20, 3949-3958.
    OpenUrlCrossRefPubMedWeb of Science
  42. ↵
    Jurukovski, V. and Simon, M. (1999). Reduced lecithin:retinol acyl transferase activity in cultured squamous cell carcinoma lines results in increased substrate-driven retinoic acid synthesis. Biochim. Biophys. Acta 1436, 479-490.
    OpenUrlPubMed
  43. ↵
    Kaneda, A., Kaminishi, M., Yanagihara, K., Sugimura, T. and Ushijima, T. (2002). Identification of silencing of nine genes in human gastric cancers. Cancer Res. 62, 6645-6650.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Li, L., Ren, C. H., Tahir, S. A., Ren, C. and Thompson, T. C. (2003). Caveolin-1 maintains activated Akt in prostate cancer cells through scaffolding domain binding site interactions with and inhibition of serine/threonine protein phosphatases PP1 and PP2A. Mol. Cell. Biol. 23, 9389-9404.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Li, X., Yost, H. J., Virshup, D. M. and Seeling, J. M. (2001). Protein phosphatase 2A and its B56 regulatory subunit inhibit Wnt signaling in Xenopus. EMBO J. 20, 4122-4131.
    OpenUrlAbstract
  46. ↵
    Li, X., Scuderi, A., Letsou, A. and Virshup, D. M. (2002). B56-associated protein phosphatase 2A is required for survival and protects from apoptosis in Drosophila melanogaster. Mol. Cell. Biol. 22, 3674-3684.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    Lotz, K., Kellner, T., Heitmann, M., Nazarenko, I., Noske, A., Malek, A., Gontarewicz, A., Schäfer, R. and Sers, C. (2005). Suppression of the TIG3 tumor suppressor gene in human ovarian carcinomas is mediated via mitogen-activated kinase-dependent and -independent mechanisms. Int. J. Cancer 116, 894-902.
    OpenUrlCrossRefPubMed
  48. ↵
    MacDonald, P. N. and Ong, D. E. (1988). Evidence for a lecithin-retinol acyltransferase activity in the rat small intestine. J. Biol. Chem. 263, 12478-12482.
    OpenUrlAbstract/FREE Full Text
  49. MacKeigen, J. P., Murphy, L. O. and Blenis, J. (2005). Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance. Nat. Cell Biol. 7, 591-600.
    OpenUrlCrossRefPubMedWeb of Science
  50. ↵
    McCluskey, A., Sim, A. T. and Sakoff, J. A. (2002). Serine-threonine protein phosphatase inhibitors: development of potential therapeutic strategies. J. Med. Chem. 45, 1151-1175.
    OpenUrlCrossRefPubMedWeb of Science
  51. ↵
    Millward, T. A., Zolnierowicz, S. and Hemmings, B. A. (1999). Regulation of protein kinase cascades by protein phosphatase 2A. Trends Biochem. Sci. 24, 186-191.
    OpenUrlCrossRefPubMedWeb of Science
  52. ↵
    Mondal, M. S., Ruiz, A., Bok, D. and Rando, R. R. (2000). Lecithin retinol acyltransferase contains cysteine residues essential for catalysis. Biochemistry 39, 5215-5220.
    OpenUrlCrossRefPubMed
  53. ↵
    Mondal, M. S., Ruiz, A., Hu, J., Bok, D. and Rando, R. R. (2001). Two histidine residues are essential for catalysis by lecithin retinol acyl transferase. FEBS Lett. 489, 14-18.
    OpenUrlCrossRefPubMedWeb of Science
  54. ↵
    Nunbhakdi-Craig, V., Machleidt, T., Ogris, E., Bellotto, D., White, C. L., III and Sontag, E. (2002). Protein phosphatase 2A associates with and regulates atypical PKC and the epithelial tight junction complex. J. Cell Biol. 158, 967-978.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    Prickett, T. D. and Brautigan, D. L. (2004). Overlapping binding sites in protein phosphatase 2A for association with regulatory A and alpha-4 (mTap42) subunits. J. Biol. Chem. 279, 38912-38920.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Ratcliffe, M. J., Itoh, K. and Sokol, S. Y. (2000). A positive role for the PP2A catalytic subunit in Wnt signal transduction. J. Biol. Chem. 275, 35680-35683.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    Ricciarelli, R. and Azzi, A. (1998). Regulation of recombinant PKC alpha activity by protein phosphatase 1 and protein phosphatase 2A. Arch. Biochem. Biophys. 355, 197-200.
    OpenUrlCrossRefPubMedWeb of Science
  58. ↵
    Ruiz, A., Winston, A., Lim, Y. H., Gilbert, B. A., Rando, R. R. and Bok, D. (1999). Molecular and biochemical characterization of lecithin retinol acyltransferase. J. Biol. Chem. 274, 3834-3841.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    Sager, R. (1997). Expression genetics in cancer: shifting the focus from DNA to RNA. Proc. Natl. Acad. Sci. USA 94, 952-955.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    Sasaki, K., Murata, M., Yasumoto, T., Mieskes, G. and Takai, A. (1994). Affinity of okadaic acid to type-1 and type-2A protein phosphatases is markedly reduced by oxidation of its 27-hydroxyl group. Biochem. J. 298, 259-262.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Schonthal, A. H. (1998). Role of PP2A in intracellular signal transduction pathways. Front. Biosci. 3, D1262-D1273.
    OpenUrlPubMed
  62. ↵
    Seeling, J. M., Miller, J. R., Gil, R., Moon, R. T., White, R. and Virshup, D. M. (1999). Regulation of beta-catenin signaling by the B56 subunit of protein phosphatase 2A. Science 283, 2089-2091.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    Sers, C., Emmenegger, U., Husmann, K., Bucher, K., Andres, A. C. and Schäfer, R. (1997). Growth-inhibitory activity and downregulation of the class II tumor-suppressor gene H-rev107 in tumor cell lines and experimental tumors. J. Cell Biol. 136, 935-944.
    OpenUrlAbstract/FREE Full Text
  64. ↵
    Sers, C., Husmann, K., Nazarenko, I., Reich, S., Wiechen, K., Zhumabayeva, B., Adhikari, P., Schroder, K., Gontarewicz, A. and Schäfer, R. (2002). The class II tumour suppressor gene H-REV107-1 is a target of interferon-regulatory factor-1 and is involved in IFNgamma-induced cell death in human ovarian carcinoma cells. Oncogene 21, 2829-2839.
    OpenUrlCrossRefPubMedWeb of Science
  65. ↵
    Shyu, R. Y., Jiang, S. Y., Chou, J. M., Shih, Y. L., Lee, M. S., Yu, J. C., Chao, P. C., Hsu, Y. J. and Jao, S. W. (2003). RARRES3 expression positively correlated to tumour differentiation in tissues of colorectal adenocarcinoma. Br. J. Cancer 89, 146-151.
    OpenUrlCrossRefPubMed
  66. ↵
    Siegrist, S., Feral, C., Chami, M., Solhonne, B., Mattei, M. G., Rajpert-De Meyts, E., Guellaen, G. and Bulle, F. (2001). hH-Rev107, a class II tumor suppressor gene, is expressed by post-meiotic testicular germ cells and CIS cells but not by human testicular germ cell tumors. Oncogene 20, 5155-5163.
    OpenUrlCrossRefPubMed
  67. ↵
    Silverstein, A. M., Barrow, C. A., Davis, A. J. and Mumby, M. C. (2002). Actions of PP2A on the MAP kinase pathway and apoptosis are mediated by distinct regulatory subunits. Proc. Natl. Acad. Sci. USA 99, 4221-4226.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    Sim, A. T. and Scott, J. D. (1999). Targeting of PKA, PKC and protein phosphatases to cellular microdomains. Cell Calcium 26, 209-217.
    OpenUrlCrossRefPubMedWeb of Science
  69. ↵
    Simmons, D. P., Andreola, F. and De Luca, L. M. (2002). Human melanomas of fibroblast and epithelial morphology differ widely in their ability to synthesize retinyl esters. Carcinogenesis 23, 1821-1830.
    OpenUrlAbstract/FREE Full Text
  70. ↵
    Sontag, E. (2001). Protein phosphatase 2A: the Trojan Horse of cellular signaling. Cell. Signal. 13, 7-16.
    OpenUrlCrossRefPubMedWeb of Science
  71. ↵
    Sontag, E., Sontag, J. M. and Garcia, A. (1997). Protein phosphatase 2A is a critical regulator of protein kinase C zeta signaling targeted by SV40 small t to promote cell growth and NF-kappaB activation. EMBO J. 16, 5662-5671.
    OpenUrlAbstract
  72. ↵
    Standaert, M. L., Bandyopadhyay, G., Sajan, M. P., Cong, L., Quon, M. J. and Farese, R. V. (1999). Okadaic acid activates atypical protein kinase C (zeta/lambda) in rat and 3T3/L1 adipocytes. An apparent requirement for activation of Glut4 translocation and glucose transport. J. Biol. Chem. 274, 14074-14078.
    OpenUrlAbstract/FREE Full Text
  73. ↵
    Strack, S., Cribbs, J. T. and Gomez, L. (2004). Critical role for protein phosphatase 2A heterotrimers in mammalian cell survival. J. Biol. Chem. 279, 47732-47739.
    OpenUrlAbstract/FREE Full Text
  74. ↵
    Sturniolo, M. T., Dashti, S. R., Deucher, A., Rorke, E. A., Broome, A. M., Chandraratna, R. A. S., Keepers, T. and Eckert, R. L. (2003). A novel tumor suppressor protein promotes keratinocyte terminal differentiation via activation of type I transglutaminase. J. Biol. Chem. 278, 48066-48073.
    OpenUrlAbstract/FREE Full Text
  75. ↵
    Tsai, F. M., Shyu, R. Y. and Jiang, S. Y. (2006). RIG1 inhibits the Ras/mitogen-activated protein kinase pathway by suppressing the activation of Ras. Cell. Signal. 18, 349-358.
    OpenUrlCrossRefPubMed
  76. ↵
    Turowski, P., Fernandez, A., Favre, B., Lamb, N. J. and Hemmings, B. A. (1995). Differential methylation and altered conformation of cytoplasmic and nuclear forms of protein phosphatase 2A during cell cycle progression. J. Cell Biol. 129, 397-410.
    OpenUrlAbstract/FREE Full Text
  77. ↵
    Wu, J., Tolstykh, T., Lee, J., Boyd, K., Stock, J. B. and Broach, J. R. (2000). Carboxyl methylation of the phosphoprotein phosphatase 2A catalytic subunit promotes its functional association with regulatory subunits in vivo. EMBO J. 19, 5672-5681.
    OpenUrlAbstract
  78. ↵
    Xue, L. and Rando, R. R. (2004). Roles of cysteine 161 and tyrosine 154 in the lecithin-retinol acyltransferase mechanism. Biochemistry 43, 6120-6126.
    OpenUrlCrossRefPubMed
  79. ↵
    Xue, L., Jahng, W. J., Gollapalli, D. and Rando, R. R. (2006). Palmitoyl transferase activity of lecithin retinol acyl transferase. Biochemistry 45, 10710-10718.
    OpenUrlCrossRefPubMedWeb of Science
  80. ↵
    Yamamoto, H., Hinoi, T., Michiue, T., Fukui, A., Usui, H., Janssens, V., Van Hoof, C., Goris, J., Asashima, M. and Kikuchi, A. (2001). Inhibition of the Wnt signaling pathway by the PR61 subunit of protein phosphatase 2A. J. Biol. Chem. 276, 26875-26882.
    OpenUrlAbstract/FREE Full Text
  81. ↵
    Yang, J., Wu, J., Tan, C. and Klein, P. S. (2003). PP2A:B56epsilon is required for Wnt/beta-catenin signaling during embryonic development. Development 130, 5569-5578.
    OpenUrlAbstract/FREE Full Text
  82. ↵
    Zhan, H. C., Gudas, L. J., Bok, D., Rando, R., Nanus, D. M. and Tickoo, S. K. (2003). Differential expression of the enzyme that esterifies retinol, lecithin:retinol acyltransferase, in subtypes of human renal cancer and normal kidney. Clin. Cancer Res. 9, 4897-4905.
    OpenUrlAbstract/FREE Full Text
  83. ↵
    Zhang, L., Huang, J., Yang, N., Liang, S., Barchetti, A., Giannakakis, A., Cadungog, M. G., O'Brien-Jenkins, A., Massobrio, M., Roby, K. F. et al. (2006). Integrative genomic analysis of protein kinase C (PKC) family identifies PKCiota as a biomarker and potential oncogene in ovarian carcinoma. Cancer Res. 66, 4627-4635.
    OpenUrlAbstract/FREE Full Text
  84. ↵
    Zhou, B., Wang, Z. X., Zhao, Y., Brautigan, D. L. and Zhang, Z. Y. (2002). The specificity of extracellular signal-regulated kinase 2 dephosphorylation by protein phosphatases. J. Biol. Chem. 277, 31818-31825.
    OpenUrlAbstract/FREE Full Text
  85. ↵
    Zuluaga, S., Alvarez-Barrientos, A., Gutierrez-Uzquiza, A., Benito, M., Nebreda, A. R. and Porras, A. (2007). Negative regulation of Akt activity by p38alpha MAP kinase in cardiomyocytes involves membrane localization of PP2A through interaction with caveolin-1. Cell. Signal. 19, 62-74.
    OpenUrlCrossRefPubMedWeb of Science
View Abstract
Previous ArticleNext Article
Back to top
Previous ArticleNext Article

This Issue

 Download PDF

Email

Thank you for your interest in spreading the word on Journal of Cell Science.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Mechanisms of the HRSL3 tumor suppressor function in ovarian carcinoma cells
(Your Name) has sent you a message from Journal of Cell Science
(Your Name) thought you would like to see the Journal of Cell Science web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Research Article
Mechanisms of the HRSL3 tumor suppressor function in ovarian carcinoma cells
Irina Nazarenko, Reinhold Schäfer, Christine Sers
Journal of Cell Science 2007 120: 1393-1404; doi: 10.1242/jcs.000018
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
Citation Tools
Research Article
Mechanisms of the HRSL3 tumor suppressor function in ovarian carcinoma cells
Irina Nazarenko, Reinhold Schäfer, Christine Sers
Journal of Cell Science 2007 120: 1393-1404; doi: 10.1242/jcs.000018

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Alerts

Please log in to add an alert for this article.

Sign in to email alerts with your email address

Article navigation

  • Top
  • Article
    • Summary
    • Introduction
    • Results
    • Discussion
    • Materials and Methods
    • Acknowledgments
    • Footnotes
    • References
  • Figures & tables
  • Supp info
  • Info & metrics
  • PDF

Related articles

Cited by...

More in this TOC section

  • Histone chaperone APLF level dictates the implantation of mouse embryos
  • Switching between blebbing and lamellipodia depends on the degree of non-muscle myosin II activity
  • Kindlin-2 promotes rear focal adhesion disassembly and directional persistence during cell migration
Show more RESEARCH ARTICLE

Similar articles

Other journals from The Company of Biologists

Development

Journal of Experimental Biology

Disease Models & Mechanisms

Biology Open

Advertisement

2020 at The Company of Biologists

Despite the challenges of 2020, we were able to bring a number of long-term projects and new ventures to fruition. While we look forward to a new year, join us as we reflect on the triumphs of the last 12 months.


Mole – The Corona Files

"This is not going to go away, 'like a miracle.' We have to do magic. And I know we can."

Mole continues to offer his wise words to researchers on how to manage during the COVID-19 pandemic.


Cell scientist to watch – Christine Faulkner

In an interview, Christine Faulkner talks about where her interest in plant science began, how she found the transition between Australia and the UK, and shares her thoughts on virtual conferences.


Read & Publish participation extends worldwide

“The clear advantages are rapid and efficient exposure and easy access to my article around the world. I believe it is great to have this publishing option in fast-growing fields in biomedical research.”

Dr Jaceques Behmoaras (Imperial College London) shares his experience of publishing Open Access as part of our growing Read & Publish initiative. We now have over 60 institutions in 12 countries taking part – find out more and view our full list of participating institutions.


JCS and COVID-19

For more information on measures Journal of Cell Science is taking to support the community during the COVID-19 pandemic, please see here.

If you have any questions or concerns, please do not hestiate to contact the Editorial Office.

Articles

  • Accepted manuscripts
  • Latest complete issue
  • Issue archive
  • Archive by article type
  • Special issues
  • Subject collections
  • Interviews
  • Sign up for alerts

About us

  • About Journal of Cell Science
  • Editors and Board
  • Editor biographies
  • Travelling Fellowships
  • Grants and funding
  • Journal Meetings
  • Workshops
  • The Company of Biologists

For Authors

  • Submit a manuscript
  • Aims and scope
  • Presubmission enquiries
  • Fast-track manuscripts
  • Article types
  • Manuscript preparation
  • Cover suggestions
  • Editorial process
  • Promoting your paper
  • Open Access
  • JCS Prize
  • Manuscript transfer network
  • Biology Open transfer

Journal Info

  • Journal policies
  • Rights and permissions
  • Media policies
  • Reviewer guide
  • Sign up for alerts

Contacts

  • Contact JCS
  • Subscriptions
  • Advertising
  • Feedback

Twitter   YouTube   LinkedIn

© 2021   The Company of Biologists Ltd   Registered Charity 277992