FKBP10/FKBP65 expression in high-grade ovarian serous carcinoma and its association with patient outcome

  • Authors:
    • Michael C.J. Quinn
    • Paulina M. Wojnarowicz
    • Amy Pickett
    • Diane M. Provencher
    • Anne-Marie Mes-Masson
    • Elaine C. Davis
    • Patricia N. Tonin
  • View Affiliations

  • Published online on: January 23, 2013     https://doi.org/10.3892/ijo.2013.1797
  • Pages: 912-920
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The frequent loss of chromosome 17 in epithelial ovarian carcinomas (EOC), particularly high-grade serous carcinomas (HGSC), has been attributed to the disruption of TP53 (at 17p13.1) and other chromosome 17 genes suspected to play a role in tumour suppressor pathways. In a transcriptome analysis of HGSC, we showed underexpression of a number of chromosome 17 genes, which included FKBP10 (at 17q21.1) and collagen I α 1 (COL1A1; at 17q21.33). FKBP10 codes for the immunophilin FKBP65 and is suspected to act as a chaperone for COL1A1. We have investigated FKBP10 (gene) and FKBP65 (protein) expression in HGSC samples and EOC cell lines that differ in their tumourigenic potential. COL1A1 expression was also investigated given the purported function of FKBP65. RT-PCR analysis verified underexpression of FKBP10 and COL1A1 in HGSCs (n=14) and six tumourigenic EOC cell lines, relative to normal ovarian surface epithelial cells and a non‑tumourigenic EOC cell line. Immunohistochemistry analyses of 196 HGSC samples using tissue microarrays revealed variable staining intensities in the epithelial tumour component where only 7.8% and 1.0% of samples stained intensely for FKBP65 and COL1A1, respectively. Variable staining intensities were also observed for the stromal component where 23.6% and 24.1% stained intensely for FKBP65 and COL1A1, respectively. There was no significant correlation of staining intensity of either protein with disease stage. Staining of FKBP65 was clearly visible in normal epithelial cells of the ovarian surface and fallopian tube. There was a significant correlation between absence of FKBP65 staining in the epithelial cell component of the tumour and prolonged overall survival (p<0.001). Our results suggest that underexpression of FKBP65 protein is characteristic of HGSCs and that this expression profile may be linked to molecular pathways associated with an unfavourable outcome in cancer patients.

Introduction

Loss of heterozygosity of chromosome 17 is a frequent occurrence in epithelial ovarian cancer (EOC) and particularly in high-grade serous carcinomas (HGSC), which is one of the most common histotypes of EOCs (16). This has been largely attributed to the inactivation of the tumour suppressor gene TP53, which is located at 17p13.1, as it is the most frequently mutated gene in HGSCs, however, other genes involved in tumour suppressor pathways have been proposed (14,714). For example, our group recently identified 158 underexpressed chromosome 17 genes in a transcriptome analysis of HGSCs, which included FKBP10, a gene that maps to 17q21.1. Interestingly, FKBP10 was among the genes upregulated in a genetically modified EOC cell line rendered non-tumourigenic as a consequence of an unique gene complementation assay involving chromosome transfer, and thus may be one of a number of genes transcriptionally reprogrammed as a consequence of tumour suppression (15,16).

FKBP10 encodes FKBP65, a 65 kDa FK506 binding protein that is a member of the FKBP-type peptidylprolyl cis/trans isomerase family (17,18). This protein localizes to the endoplasmic reticulum and acts as a molecular chaperone and binding partner of type I collagen (1921). The interaction with type I collagen is interesting as COL1A1, which is a gene that maps to 17q21.33, was also found significantly underexpressed in our chromosome 17 transcriptome analyses of HGSCs (14). FKBP65 is expressed in developing tissues and re-expressed in adult tissues following injury (19,20). In the mouse, the only tissues that continue to express FKBP65 are reproductive tissues (ovary, uterus and mammary glands) during phases of growth and remodelling (E.C. Davis, unpublished data). Although somatic mutations inactivating FKBP10 have not been identified, such as in recent reports of genome-wide exomic sequencing analyses of HGSCs by The Cancer Genome Atlas Research Network (5) and other cancer types (22), germline FKBP10 mutations have been described in association with autosomal-recessive osteogenesis imperfecta and Bruck syndrome, which are connective tissue disorders characterized by defects in type I collagen (23,24). Though epithelial malignancies in patients with osteogenesis imperfecta are rare, there is at least one case report of a 32-year-old who developed a low-grade serous ovarian carcinoma with stage IIIb disease (25).

The localization of FKBP10 to chromosome 17 and its interesting expression profile in the murine adult normal ovary, HGSCs and our genetically modified EOC cell line, prompted our further investigation of this gene in ovarian cancer samples. In this study, we investigated the expression profile of FKBP10 in HGSCs, and its expression in a set of well-characterized EOC cell lines that differ in their growth characteristics and tumourigenic potential. We also investigated protein expression by immunohistochemistry analysis of a tissue microarray and related the expression profile to patient outcome. COLIA1 gene and protein expression was also investigated given its purported interaction with FKBP65.

Materials and methods

Tissue specimens, cell lines, and clinical information

The HGSCs, primary cultures of normal ovarian surface epithelial cells (NOSE), and EOC cell lines (OV90, TOV112D, TOV81D, TOV21G, TOV1946, OV1946 and TOV2223) examined for gene expression have been described previously (1,14,26). Briefly, the HGSC samples were from chemotherapy naïve patients and the cell lines were derived from long-term passages of malignant ovarian ascites from an undifferentiated adeno-carcinoma (OV-90), high-grade endometrioid adenocarcinoma (TOV112D), serous carcinomas (TOV81D, TOV1946, OV1946 and TOV2223) and a clear cell carcinoma (TOV21G), where TOV1946 and OV1946 were derived from malignant ovarian ascites (OV1946) or tumour (TOV1946) from the same patient.

The HGSC (n=196) cases represented in the tissue array were derived from archival blocks of paraffin-embedded tissues samples as previously described (27). The tumour grade and disease stage (Table I) were designated according to the International Federation of Gynaecology and Obstetrics. Disease-free interval, defined as time to doubling of the upper normal limit of the serum cancer antigen marker CA-125 or the detection of a new lesion by ultrasound or CT-scan imaging, and overall survival, defined according to the Response Evaluation Criteria in Solid Tumors (28) were extracted from the Système d’Archivage des Données en Oncologie (Table I). Normal ovary and fallopian tube tissues used in immunohistochemistry analyses were retrieved from archival paraffin-embedded samples. Normal ovarian tissues were obtained from ovarectomy (age 40) and hysterectomy (age 45) cases due to a benign uterine tumour. The fallopian tube tissues were adjacent normal tissues collected from women diagnosed with serous ovarian cancer. All samples and related clinical information were obtained with informed written consent at the Centre Hospitalier de l’ Universite de Montreal (CHUM) - Hôpital Notre Dame.

Table I

Description of HGSC tissue array.

Table I

Description of HGSC tissue array.

Patient characteristicAll casesG2G3
No. of cases19630166
Mean age of diagnosis, years (range)62 (34–89)60 (42–82)62 (34–89)
Stage I15213
Stage II19217
Stage III13923116
Stage IV23320
Mean disease free interval, months (range)22 (0–134)15 (0–57)23 (0–134)
No. of censured patients52646
No. of non-censured patients14424120
Mean overall survival, months (range)35 (0–134)28 (0–102)36 (0–134)
No. of censured patients1101991
No. of non-censured patients861175
Gene expression analyses

Gene expression was assessed by semi-quantitative RT-PCR analysis using cDNA synthesized from total RNA prepared as previously described (3,13,15,29). Primers were designed using Primer3 software (30) based on genomic structures of FKBP10 and COL1A1 available from the March 2006 human reference sequence (NCBI Build 36.1/hg18) assembly (31) and alignment of reference sequences of each gene, NM_021939 and NM_000088.3, respectively. The FKBP10 forward primer is 5′-GTGGAACAAGGAAGA CACC-3′, and the reverse primer is 5′-CTTCCTTCTCTCTCC AGGAC-3′, yielding a 238 base pair product. The COL1A1 forward primer is 5′-GTGCTCCTGTATTGCTG-3′, and the reverse primer is 5′-CTCGCTTTCCTTCCTCTC-3′, yielding a 207 base pair product. The RT-PCR-based assays were performed essentially as previously described (32). RT-PCR of 18S RNA was performed to assess RNA quality. Primer sequences for 18S were reported previously (3).

Immunohistochemistry analysis of tissue arrays

FKBP65 and COL1A1 protein expression was assessed by immunohisto-chemistry analysis using a tissue array containing 0.6 mm cores derived from paraffin-embedded tissue blocks representing 196 HGSC cases selected based on a review of hematoxylin and eosin-stained slides prepared as described previously (27). The array also contained 11 normal fallopian tube samples. Immunohistochemistry analysis was performed on the tissue array and sections prepared from two normal ovary paraffin-embedded tissue blocks. Five-micron sections were mounted onto frosted plus slides, deparaffinized in Citrisolv (Fisher Scientific) for 90 min and then rehydrated in an ethanol gradient. Before primary antibody incubation, antigen retrieval was performed using 0.05% Tween-20 in 10 mM sodium citrate (pH 6.0) at 90°C for 20 min. The slides were washed with 0.1% Triton X-100 in Tris-buffered saline (TBS) [0.5 M Tris, 1.5 M NaCl, (pH 7.4)]. The slides were then washed and blocked with TBS containing 0.1% bovine serum albumin (TBS/BSA) for 3×10 min and then incubated with Ultra V Block (LabVision Corp., Fremont, CA, USA) for 7 min. After washing again in TBS/BSA, the slides were incubated overnight at 4°C with either a polyclonal FKBP65 antibody, raised against a synthetic peptide to the C-terminus of the mouse FKBP65 (33), or a polyclonal type I collagen antibody (Calbiochem), diluted at 1:300 and 1:1000, respectively. Slides were incubated in Value Primary Antibody Enhancer (LabVision Corp.) for 20 min before incubation in AP Value Polymer (LabVision Corp.) for 30 min at room temperature. Staining was visualized using FastRed (LabVision Corp.) and slides were counterstained with hematoxylin for 30 sec. The tissue arrays were scanned with an Aperio ScanScope XT Digital Slide Scanner and images were viewed at high resolution using Aperio ImageScope Software version 11.02. Two observers examined the images independently and scored them based on staining intensity ranked as absent, low, moderate or high, for both the epithelial and stromal tissue components of each core. The inter-observer correlation coefficients were 0.792 for FKBP65 and 0.747 for COL1A1. The inter-observer correlation coefficient was calculated using SPSS software version 16.0 (SPSS Inc., Chicago, IL, USA), where the minimum threshold was 0.7.

Statistical analyses

Spearman analysis was used to evaluate the correlation of FKBP65 and COL1A1 staining intensities. The association between tumour grade and staining intensity was evaluated using an independent sample t-test. The association between disease stage and staining intensity was evaluated using a one-way ANOVA. The relationship between staining intensity and disease-free interval or overall survival was evaluated using the log-rank test and visualized by Kaplan-Meier survival curve analysis. All statistical analyses were performed with SPSS software version 16.0 (SPSS Inc.), p-values <0.05 were considered significant.

Results

Gene expression analyses of FKBP10 and COL1A1

A previous gene expression microarray analysis of chromo-some 17 genes identified FKBP10 and COL1A1 among the 158 genes underexpressed in HGSCs as compared with NOSEs (14). To verify these observations, we investigated the expression of these genes by performing RT-PCR analyses on the samples used in the microarray analyses. As shown in Fig. 1, FKBP10 and COL1A1 expression was clearly detectable in all NOSEs. In contrast, FKBP10 expression in the HGSCs was undetectable or expressed at levels lower than that observed in the NOSEs. COL1A1 expression was more variable, ranging from clearly detectable levels comparable to those observed in NOSEs in some samples, to undetectable or lower levels of expression relative to the NOSEs (Fig. 1). Gene expression of FKBP10 and COL1A1 was also investigated in cell lines established as long-term passages from chemotherapy naïve EOC samples (1,26). FKBP10 expression was detectable in all EOC cell lines with the highest level of expression observed in TOV81D (Fig. 1). In contrast, COL1A1 expression was detectable in only TOV81D (Fig. 1), which is a non-tumourigenic EOC cell line.

Immunohistochemical staining of FKBP65 and COL1A1

Immunohistochemistry analysis was performed to characterize FKBP65 protein expression in normal ovary and fallopian tube as the expression profile in human tissues purported to be the origins of HGSCs have not previously been described. Staining was evident in both the epithelial and stromal cells of the tissues (Fig. 2).

FKBP65 and COL1A1 protein expression in HGSCs was investigated by immunohistochemistry analysis using a tissue array containing 196 cores from tumour samples (Table I). Staining was localized to the cytoplasm for both FKBP65 and COL1A1 and was observed in both the epithelial and stromal cell components of the tumour samples (Fig. 3). To characterize the expression pattern, intensity of staining was scored as absent, low, moderate or high for both the epithelial and stromal cell components where possible, as scoring was not possible for some of the samples due to the type of cells present or the quality of core (Fig. 4). The majority of the epithelial cell components of the tumour samples scored as either low or moderate for FKBP65 expression, and less than 10% of samples scored as either absent or high (Table II). A somewhat similar staining intensity pattern for the epithelial component of the tumours was also observed with COL1A1, although there were more cases that scored with low intensity levels (Table II). In contrast, FKBP65 expression patterns for the stromal components of the tumour were almost equally distributed among all staining intensity categories, with the largest number of samples (32.3%) exhibiting moderate staining intensity (Table II). Although the largest number of samples (44.5%) also exhibited moderate staining of COL1A1 in the stromal component of the tumour, there were fewer samples that had an absent intensity score (Table II). There was a significant correlation between FKBP65 and COL1A1 staining intensity in the epithelial cell component (p<0.001), but not in the stromal component (p=0.101) of the tumour samples analysed.

Table II

Distribution of staining intensities for FKBP65 and COL1A1 in HGSC samples.

Table II

Distribution of staining intensities for FKBP65 and COL1A1 in HGSC samples.

ProteinCell typeTotal no. of samples scoredStaining intensity score (%)

AbsentLowModerateHigh
FKBP65Epithelial1932 (1.0)107 (55.4)69 (35.8)15 (7.8)
Stromal19546 (23.6)40 (20.5)63 (32.3)46 (23.6)
COL1A1Epithelial1912 (1.0)134 (70.2)53 (27.7)2 (1.0)
Stromal1919 (4.7)51 (26.7)85 (44.5)46 (24.1)
Protein expression and disease stage, disease-free interval or overall survival

The staining intensity patterns of FKBP65 and COL1A1 were characterized with respect to disease stage, though the majority of samples (83%) were from cases with advanced stage III/IV disease (Table I). There were no statistically significant differences (data not shown) in the distribution of staining intensity patterns for the proteins assayed for either the epithelial or stromal cell components of the tumour samples and disease stage (Table III).

Table III

Distribution of staining intensities for FKBP65 and COL1A1 by stage.

Table III

Distribution of staining intensities for FKBP65 and COL1A1 by stage.

ProteinCell typeStageTotal no. of samples scoredStaining intensity score (%)

AbsentLowModerateHigh
FKBP65EpithelialI1509 (60)5 (33.3)1 (6.6)
II18012 (66.6)5 (27.7)1 (5.5)
III1372 (1.5)75 (54.7)52 (38.0)8 (5.8)
IV23011 (97.8)7 (30.4)5 (21.7)
StromalI155 (33.3)2 (13.3)4 (26.6)4 (26.6)
II1906 (31.6)8 (42.1)5 (26.3)
III13837 (26.8)27 (19.6)42 (30.4)32 (23.2)
IV234 (17.4)5 (21.7)9 (39.1)5 (21.7)
COL1A1EpithelialI15010 (66.6)5 (33.3)0
II19015 (78.9)4 (21.1)0
III1351 (0.7)98 (72.6)36 (26.7)0
IV221 (4.5)11 (50)8 (36.4)2 (9.1)
StromalI151 (6.6)2 (13.3)6 (40)6 (40)
II1805(27.8)7 (38.9)6 (33.3)
III1366 (4.4)37(27.2)65 (47.8)28(20.6)
IV222 (9.1)7 (31.8)7 (31.8)6 (27.3)

The staining intensities for FKBP65 and COL1A1 were also evaluated with respect to disease-free interval and overall survival. No significant relationships were observed with staining intensities of COL1A1 for the epithelial or stromal components of the tumour samples and either of these clinical parameters (data not shown). Significant relationships were also not found with staining intensities of FKBP65 for the epithelial or stromal components of the tumour samples and disease-free interval (data not shown). In contrast, there was a significant association between prolonged overall survival and the presence of FKBP65 protein in the epithelial component when evaluated for each staining category (p=0.005) or when cases with no staining are compared with those with low, moderate and high staining combined (p<0.001) (Fig. 5a and c). Although there are only two samples with no detectable staining in the epithelial component of the tumour, it is interesting that the cases with the highest staining levels for FKBP65 where among the cases which had the longest overall survival (Fig. 5c). A similar analysis of FKBP65 staining found a significant association (p=0.027) with prolonged overall survival when the samples were analysed for the absence or presence of staining in the stromal component (Fig. 5b). Although there was no significant difference with overall survival and staining based on each staining intensity category (p=0.087), it is interesting that the cases scored absent for staining in the stromal component were among those with the poorest overall survival (Fig. 5d).

Discussion

In this study we have verified that FKBP10 expression is absent or low in HGSC and the results are consistent with the low frequency of high intensity staining patterns of FKBP65 in tumour samples. FKBP65 expression in the epithelial cells of normal ovarian surface and the distal fimbrae of the fallopian tube is interesting considering that both of these tissues have been proposed as the progenitor cell type for HGSC (34). During the course of this study, decreased expression of FKBP65 in a study of 57 EOC samples of different histological subtypes, which included HGSCs, was reported independently (35). These observations suggest the possibility that FKBP65 expression is also important in the biology of the other histological subtypes of EOC. This notion is supported by the observation that FKBP10 was underexpressed in most of our EOC cell lines, which were derived from epithelial ovarian tumour samples that differed in their histology.

Our results from the FKBP10 expression assays of the EOC cell lines also suggest that underexpression might be associated with tumourigenicity. Although our assays were semi-quanti tative, the highest level of gene expression was observed in TOV81D, the only cell line in our series of EOC cell lines tested that is unable to form tumours in mouse tumour xenograft assays and lacks in vitro growth pheno-types characteristics of tumourigenic cell lines (26,36). Our expression results are consistent with previously published studies from our group where, by semi-quantitative RT-PCR and gene expression microarray analyses, TOV81D exhibited gene expression profiles that resemble those of NOSEs (3,14,16,3739). Notable also is the underexpression of FKBP10 observed in the tumourigenic OV90 cell line, as this gene was induced in OV90 cell line hybrids that were rendered nontumourigenic as a consequence of the transfer chromosome 3 in genetic complementation assays aimed at identifying genes implicated in tumour suppressor pathways (15,16). Thus, decreased expression of FKBP10 may be important in tumour suppressor pathways.

We observed a significant association between FKBP65 staining intensity in the epithelial cells of HGSC samples and overall survival. An association between high FKBP65 expression and prolonged survival was also observed in the independent study of 57 EOC samples by Henriksen et al, although the finding was not significant (35). In their association analyses the small sample size and possibly the inclusion of different histological subtypes, which are known to exhibit differences in outcome (40), may have affected the interpretation of results. In our analyses, though the samples with no FKBP65 staining exhibited the poorest outcome, they represented only two HGSC cases in our cohort. Notable, however, is that the samples exhibiting the highest staining intensity were among the HGSC cases with the longest overall survival. Interestingly, the staining intensities of the stromal component of the tumour samples were also associated with overall survival. There is mounting evidence that ovarian cancer progression involves reciprocal communication between malignant epithelial cells and the adjacent stromal microenvironment possibly relating to malignant epithelium-activated fibroblasts (41). Although not fully explored in the context of ovarian cancer, gene expression profiles of stromal cells have been found to be predictive of disease outcome in breast cancer (42). The expression profile of COL1A1 was similar to FKBP65 as demonstrated by the significant positive correlation of staining intensities by immunohistochemistry of HGSCs. The similarities in expression profiles have been reported previously, and this is consistent with FKBP65 being a type I collagen chaperone (19,21).

In conclusion, we found FKBP65 underexpressed in a proportion of HGSCs, and its expression correlated with that of COL1A1 in epithelial cells. We also reported on the interesting finding that absence of FKBP65 staining was associated with poorer overall survival warranting replication of our study with larger cohorts. The interesting FKBP10 expression profiles in our EOC cell lines that differ in their tumourigenic potential could also be explored to further elucidate the molecular pathways that involve FKBP65.

Acknowledgements

We thank Jason Madore for his technical assistance. P.M.W. is a recipient of a Doctoral Research Award (DRA) from the Canadian Institute of Health Research (CIHR). The Research Institute of the McGill University Health Centre and the Centre de Recherche du Centre Hospitalier de l’ Université de Montréal receive support from the Fonds de Recherche du Québec - Santé. Clinical specimens were provided by the Banque de tissus et de données of the Réseau de Recherche sur le Cancer of the Fonds de Recherche du Québec - Santé which is affiliated with the Canadian Tumour Repository Network. This research was supported by grants from the CIHR to P.N.T., D.M.P. and A.-M.M.-M.

References

1 

Dion F, Mes-Masson AM, Seymour RJ, Provencher D and Tonin PN: Allelotyping defines minimal imbalance at chromosomal region 17q25 in non-serous epithelial ovarian cancers. Oncogene. 19:1466–1472. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Foulkes WD, Black DM, Stamp GW, Solomon E and Trowsdale J: Very frequent loss of heterozygosity throughout chromosome 17 in sporadic ovarian carcinoma. Int J Cancer. 54:220–225. 1993. View Article : Google Scholar : PubMed/NCBI

3 

Presneau N, Dewar K, Forgetta V, Provencher D, Mes-Masson AM and Tonin PN: Loss of heterozygosity and transcriptome analyses of a 1.2 Mb candidate ovarian cancer tumor suppressor locus region at 17q25.1–q25.2. Mol Carcinog. 43:141–154. 2005.PubMed/NCBI

4 

Wojnarowicz PM, Provencher DM, Mes-Masson AM and Tonin PN: Chromosome 17q25 genes, RHBDF2 and CYGB, in ovarian cancer. Int J Oncol. 40:18656–1880. 2012.PubMed/NCBI

5 

TCGA: Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar

6 

Gorringe KL, George J, Anglesio MS, et al: Copy number analysis identifies novel interactions between genomic loci in ovarian cancer. PloS One. 5:e114082010. View Article : Google Scholar : PubMed/NCBI

7 

Pieretti M, Cavalieri C, Conway PS, Gallion HH, Powell DE and Turker MS: Genetic alterations distinguish different types of ovarian tumors. Int J Cancer. 64:434–440. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Sangha N, Wu R, Kuick R, et al: Neurofibromin 1 (NF1) defects are common in human ovarian serous carcinomas and co-occur with TP53 mutations. Neoplasia. 10:1362–1372. 2008.PubMed/NCBI

9 

Feng Q, Deftereos G, Hawes SE, et al: DNA hypermethylation, Her-2/neu overexpression and p53 mutations in ovarian carcinoma. Gynecol Oncol. 111:320–329. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Rathi A, Virmani AK, Schorge JO, et al: Methylation profiles of sporadic ovarian tumors and nonmalignant ovaries from high-risk women. Clin Cancer Res. 8:3324–3331. 2002.PubMed/NCBI

11 

Pergolizzi R, Appierto V, Crosti M, et al: Role of retinoic acid receptor overexpression in sensitivity to fenretinide and tumorigenicity of human ovarian carcinoma cells. Int J Cancer. 81:829–834. 1999. View Article : Google Scholar : PubMed/NCBI

12 

Bruening W, Prowse AH, Schultz DC, Holgado-Madruga M, Wong A and Godwin AK: Expression of OVCA1, a candidate tumor suppressor, is reduced in tumors and inhibits growth of ovarian cancer cells. Cancer Res. 59:4973–4983. 1999.PubMed/NCBI

13 

Presneau N, Mes-Masson AM, Ge B, Provencher D, Hudson TJ and Tonin PN: Patterns of expression of chromosome 17 genes in primary cultures of normal ovarian surface epithelia and epithelial ovarian cancer cell lines. Oncogene. 22:1568–1579. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Wojnarowicz PM, Breznan A, Arcand SL, et al: Construction of a chromosome 17 transcriptome in serous ovarian cancer identifies differentially expressed genes. Int J Gynecol Cancer. 18:963–975. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Cody NA, Ouellet V, Manderson EN, et al: Transfer of chromosome 3 fragments suppresses tumorigenicity of an ovarian cancer cell line monoallelic for chromosome 3p. Oncogene. 26:618–632. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Quinn MC, Filali-Mouhim A, Provencher DM, Mes-Masson AM and Tonin PN: Reprogramming of the transcriptome in a novel chromosome 3 transfer tumor suppressor ovarian cancer cell line model affected molecular networks that are characteristic of ovarian cancer. Mol Carcinog. 48:648–661. 2009. View Article : Google Scholar

17 

Coss MC, Winterstein D, Sowder RC II and Simek SL: Molecular cloning, DNA sequence analysis, and biochemical characterization of a novel 65-kDa FK506-binding protein (FKBP65). J Biol Chem. 270:29336–29341. 1995. View Article : Google Scholar : PubMed/NCBI

18 

Zeng B, MacDonald JR, Bann JG, et al: Chicken FK506-binding protein, FKBP65, a member of the FKBP family of peptidylprolyl cis-trans isomerases, is only partially inhibited by FK506. Biochem J. 330:109–114. 1998.PubMed/NCBI

19 

Patterson CE, Abrams WR, Wolter NE, Rosenbloom J and Davis EC: Developmental regulation and coordinate reexpression of FKBP65 with extracellular matrix proteins after lung injury suggest a specialized function for this endoplasmic reticulum immunophilin. Cell Stress Chaperones. 10:285–295. 2005. View Article : Google Scholar

20 

Patterson CE, Schaub T, Coleman EJ and Davis EC: Developmental regulation of FKBP65. An ER-localized extra-cellular matrix binding-protein. Mol Biol Cell. 11:3925–3935. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Ishikawa Y, Vranka J, Wirz J, Nagata K and Bachinger HP: The rough endoplasmic reticulum-resident FK506-binding protein FKBP65 is a molecular chaperone that interacts with collagens. J Biol Chem. 283:31584–31590. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Forbes SA, Bindal N, Bamford S, et al: COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acid Res. 39:D945–D950. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Alanay Y, Avaygan H, Camacho N, et al: Mutations in the gene encoding the RER protein FKBP65 cause autosomal-recessive osteogenesis imperfecta. Am J Hum Genet. 86:551–559. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Kelley BP, Malfait F, Bonafe L, et al: Mutations in FKBP10 cause recessive osteogenesis imperfecta and Bruck syndrome. J Bone Miner Res. 26:666–672. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Nishida T, Oda T, Sugiyama T, Izumi S and Yakushiji M: Concurrent ovarian serous carcinoma and osteogenesis imperfecta. Arch Gynecol Obstet. 253:153–156. 1993. View Article : Google Scholar : PubMed/NCBI

26 

Ouellet V, Zietarska M, Portelance L, et al: Characterization of three new serous epithelial ovarian cancer cell lines. BMC Cancer. 8:1522008. View Article : Google Scholar : PubMed/NCBI

27 

Le Page C, Marineau A, Bonza PK, et al: BTN3A2 expression in epithelial ovarian cancer is associated with higher tumor infiltrating T cells and a better prognosis. PloS One. 7:e385412012.PubMed/NCBI

28 

Therasse P, Arbuck SG, Eisenhauer EA, et al: New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 92:205–216. 2000. View Article : Google Scholar

29 

Ouellet V, Provencher DM, Maugard CM, et al: Discrimination between serous low malignant potential and invasive epithelial ovarian tumors using molecular profiling. Oncogene. 24:4672–4687. 2005. View Article : Google Scholar

30 

Rozen S and Skaletsky H: Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol. 132:365–386. 2000.PubMed/NCBI

31 

Karolchik D, Baertsch R, Diekhans M, et al: The UCSC Genome Browser Database. Nucleic Acid Res. 31:51–54. 2003. View Article : Google Scholar

32 

Arcand SL, Maugard CM, Ghadirian P, et al: Germline TP53 mutations in BRCA1 and BRCA2 mutation-negative French Canadian breast cancer families. Breast Cancer Res Treat. 108:399–408. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Davis EC, Broekelmann TJ, Ozawa Y and Mecham RP: Identification of tropoelastin as a ligand for the 65-kD FK506-binding protein, FKBP65, in the secretory pathway. J Cell Biol. 140:295–303. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Salvador S, Gilks B, Kobel M, Huntsman D, Rosen B and Miller D: The fallopian tube: primary site of most pelvic high-grade serous carcinomas. Int J Gynecol Cancer. 19:58–64. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Henriksen R, Sorensen FB, Orntoft TF and Birkenkamp-Demtroder K: Expression of FK506 binding protein 65 (FKBP65) is decreased in epithelial ovarian cancer cells compared to benign tumor cells and to ovarian epithelium. Tumour Biol. 32:671–676. 2011. View Article : Google Scholar

36 

Provencher DM, Lounis H, Champoux L, et al: Characterization of four novel epithelial ovarian cancer cell lines. In Vitro Cell Dev Biol Anim. 36:357–361. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Birch AH, Quinn MC, Filali-Mouhim A, Provencher DM, Mes-Masson AM and Tonin PN: Transcriptome analysis of serous ovarian cancers identifies differentially expressed chromosome 3 genes. Mol Carcinog. 47:56–65. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Cody NA, Shen Z, Ripeau JS, et al: Characterization of the 3p12.3-pcen region associated with tumor suppression in a novel ovarian cancer cell line model genetically modified by chromosome 3 fragment transfer. Mol Carcinog. 48:1077–1092. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Manderson EN, Birch AH, Shen Z, Mes-Masson AM, Provencher D and Tonin PN: Molecular genetic analysis of a cell adhesion molecule with homology to L1CAM, contactin 6, and contactin 4 candidate chromosome 3p26pter tumor suppressor genes in ovarian cancer. Int J Gynecol Cancer. 19:513–525. 2009. View Article : Google Scholar

40 

Kobel M, Kalloger SE, Boyd N, et al: Ovarian carcinoma subtypes are different diseases: implications for biomarker studies. PLoS Med. 5:e2322008. View Article : Google Scholar : PubMed/NCBI

41 

Schauer IG, Sood AK, Mok S and Liu J: Cancer-associated fibroblasts and their putative role in potentiating the initiation and development of epithelial ovarian cancer. Neoplasia. 13:393–405. 2011.PubMed/NCBI

42 

Finak G, Bertos N, Pepin F, et al: Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 14:518–527. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 42 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Quinn MC, Wojnarowicz PM, Pickett A, Provencher DM, Mes-Masson A, Davis EC and Tonin P : FKBP10/FKBP65 expression in high-grade ovarian serous carcinoma and its association with patient outcome. Int J Oncol 42: 912-920, 2013
APA
Quinn, M.C., Wojnarowicz, P.M., Pickett, A., Provencher, D.M., Mes-Masson, A., Davis, E.C., & Tonin, P. . (2013). FKBP10/FKBP65 expression in high-grade ovarian serous carcinoma and its association with patient outcome. International Journal of Oncology, 42, 912-920. https://doi.org/10.3892/ijo.2013.1797
MLA
Quinn, M. C., Wojnarowicz, P. M., Pickett, A., Provencher, D. M., Mes-Masson, A., Davis, E. C., Tonin, P. ."FKBP10/FKBP65 expression in high-grade ovarian serous carcinoma and its association with patient outcome". International Journal of Oncology 42.3 (2013): 912-920.
Chicago
Quinn, M. C., Wojnarowicz, P. M., Pickett, A., Provencher, D. M., Mes-Masson, A., Davis, E. C., Tonin, P. ."FKBP10/FKBP65 expression in high-grade ovarian serous carcinoma and its association with patient outcome". International Journal of Oncology 42, no. 3 (2013): 912-920. https://doi.org/10.3892/ijo.2013.1797