Purpose: To identify molecular alterations potentially involved in predisposition to adnexal serous carcinoma (SerCa) in the nonmalignant fallopian tube epithelium (FTE) of BRCA1/2 mutation carriers, given recent evidence implicating the distal FTE as a common source for SerCa.

Experimental Design: We obtained and compared gene expression profiles of laser capture microdissected nonmalignant distal FTE from 12 known BRCA1/2 mutation carriers (FTEb) and 12 control women (FTEn) during the luteal and follicular phase, as well as 13 high-grade tubal and ovarian SerCa.

Results: Gene expression profiles of tubal and ovarian SerCa specimens were indistinguishable by unsupervised cluster analysis and significance analysis of microarrays. FTEb samples as a group, and four individual FTEb samples from the luteal phase in particular, clustered closely with SerCa rather than normal control FTE. Differentially expressed genes from these four samples relative to other FTEb samples, as well as differentially expressed genes in all FTEb luteal samples relative to follicular samples, were mapped to the I2D protein-protein interaction database, revealing a complex network affecting signaling pathways previously implicated in tumorigenesis. Two candidates, disabled homolog 2 mitogen-responsive phosphoprotein (DAB2) and Ski-like (SKIL), were further validated by real-time reverse transcription–PCR and tissue arrays. FTEb luteal and SerCa samples expressed higher levels of oncogenic SKIL and decreased levels of tumor suppressor DAB2, relative to FTEb follicular samples.

Conclusions: These findings support a common molecular pathway for adnexal SerCa and implicate factors associated with the luteal phase in predisposition to ovarian cancer in BRCA mutation carriers.

High-grade serous carcinoma (SerCa) is the most common histologic type of both ovarian and fallopian tube carcinoma, presents at an advanced stage of disease, and has a high mortality rate. Approximately 15% of SerCa cases are considered hereditary, arising in association with a germline mutation in the BRCA1 or BRCA2 genes (1, 2), which confer an estimated 50% to 60% or 18% to 23% lifetime risk, respectively (3, 4).

Until recently, no reproducible histologic cancer precursor of high-grade SerCa had been identified (57), but the unexpected finding of occult SerCa in the fallopian tubes of BRCA mutation carriers undergoing prophylactic surgery has led to the description of putative cancer precursors in the distal fallopian tube epithelium (FTE; refs. 811). These discoveries have led investigators to postulate that the distal FTE is an alternative source of SerCa designated as of ovarian origin by pathologists (1113). Comparison of SerCa and coexisting tubal intraepithelial carcinomas has shown identical mutations of the tumor suppressor p53 in each lesion, indicating that mutation of p53, a frequent event in SerCa, occurs before the development of invasive disease and supports the idea that they are causally related (14). Although it is not possible to definitively prove the tubal origin of tubal intraepithelial carcinomas, these are likely primary lesions given the previously observed resistance of tubal mucosa to direct implantation (14, 15). This concept is further supported by the finding that gene expression profiles of ovarian SerCa are more similar to normal FTE than the ovarian surface epithelium (16), as well as by the similar risk factors (17, 18) and genomic alterations of ovarian and fallopian tube cancers (19, 20).

To identify molecular alterations that may be involved in predisposition to SerCa in the nonmalignant FTE of BRCA1/2 mutation carriers, we generated and compared gene expression profiles of laser capture microdissected epithelial cells from the nonmalignant distal fallopian tube of women at a baseline risk for SerCa (FTEn) and known BRCA1/2 mutation carriers (FTEb), as well as from high-grade tubal and ovarian SerCa specimens (fallopian tube SerCa and ovarian SerCa, respectively). To our knowledge, this is the only study to date of gene expression profiles in the histologically normal FTE of BRCA1/2 mutation carriers and control patients.

Study samples. The study was approved by the University Health Network Research Ethics Board, and all patients provided informed consent. Snap-frozen tissues were selected from the University Health Network Ovarian Tissue Bank and Database, including histologically normal fallopian tubes from 12 BRCA mutation carriers (FTEb) and from 12 control women undergoing salpingo-oophorectomy for reasons other than adnexal malignancy or family history (FTEn). Consequently, these women had not been tested for the presence of a BRCA1/2 mutation. All women were premenopausal, and patients undergoing treatment for breast carcinoma, on hormonal therapy, or with a diagnosis of endometrial carcinoma were excluded. Although 3 of 12 mutation carriers had a history of breast carcinoma, they were not taking antiestrogenic therapy at the time of surgery, with an average time since last treatment of 45.7 mo [95% confidence interval (95% CI), 6.6-84.7]. Nonmalignant FTEn and FTEb specimens were age-matched, with an average age of 44.5 y (95% CI, 42.3-46.7) for control patients and 41.3 y (95% CI, 39.4-43.2) for mutation carriers. Each group of FTE included six samples obtained during the luteal phase and six from the follicular phase of the ovarian cycle, determined after review of histologic sections of ovaries and endometrium by a gynecologic pathologist (P.A.S.), to account for potential effects of the endocrine milieu on gene expression.

Thirteen SerCa samples designated as of either tubal (n = 6) or ovarian (n = 7) origin from 11 patients were included. Two independent samples were obtained from two of the patients for direct comparison of tubal and ovarian SerCa; one of these patients did not have a known family history or BRCA1/2 mutation. The remaining five ovarian SerCa patients had either a strong family history or identified mutation, whereas two of four remaining tubal SerCa patients fulfilled these criteria. The other two of four tubal SerCa patients did not have a documented family history or identified mutation, but were selected based on a similar age of cancer onset as the ovarian cases. Overall, tubal and ovarian specimens were age-matched, with an average age of 54.8 y (95% CI, 49.4-60.3) and 49.6 y (95% CI, 42.6-56.5), respectively. In addition, 12 of 13 of the SerCa specimens showed accumulation of p53 protein by immunohistochemistry (not shown). Relevant clinical information on all study patients is summarized in Table 1.

Table 1.

Relevant clinical data for samples used for gene expression profiling

SampleAgeIndication for surgery/diagnosisOvarian cycle statusBRCA1/2 statusFamily historyPrevious breast cancer
FTEn       
    1 46 History of cervical carcinoma Luteal NA No No 
    2 46 Myometrial leiomyoma Luteal NA No No 
    3 42 Grade 3 invasive squamous cell carcinoma of cervix; adenomyosis Luteal NA No No 
    4 36 Adenocarcinoma in situ of endocervix Luteal NA No No 
    5 43 Left ovary serous cystadenoma; adenomyosis Luteal NA No No 
    6 47 Uterus leiomyoma Luteal NA No No 
    7 41 Retriperitoneal high grade leiomyosarcoma Follicular NA No No 
    8 42 Grade 1 invasive adenocarcinoma of cervix; adenomyosis Follicular NA No No 
    9 46 NDH Follicular NA No No 
    10 47 History of cervical carcinoma Follicular NA No No 
    11 50 History of cervical carcinoma Follicular NA No No 
    12 48 Myometrial and subserosal uterine leiomyoma Follicular NA No No 
FTEb       
    13 44 Bilateral ovaries multiple follicular cysts; leiomyomata Luteal BRCA1 5382insC Yes No 
    14 45 Myometrial leiomyoma Luteal BRCA1 185delAG Yes No 
    15 38 NDH Luteal BRCA1 A1959T Yes No 
    16 40 Bilateral ovarian endometriosis; myometrium leiomyoma Luteal BRCA1 185delAG Unknown No 
    17 39 NDH Luteal BRCA1 185delAG Yes No 
    18 46 Right ovary cortical epithelial inclusions with hyperplasia and serous tubal metaplasia; left ovary serous cyst Luteal BRCA2 C5910G Yes No 
    19 42 Myometrium leiomyoma Follicular BRCA1 5382insC Yes Yes 
    20 40 Right ovary mature cystic teratoma negative for malignancy Follicular BRCA1 185delAG Yes No 
    21 47 Leiomyomata Follicular BRCA2 6174delT Yes No 
    22 39 Bilateral ovaries cystic follicles negative for malignancy Follicular BRCA1 1293del40 Yes No 
    23 37 NDH Follicular BRCA1 185delAG Unknown Yes 
    24 39 NDH Follicular BRCA1 185delAG Yes Yes 
Fallopian tube SerCa       
    25 41 Right tube poorly differentiated carcinoma favor serous type, grade 3; bilateral ovaries negative for malignancy NA Negative No No 
    26* 57 Left tube grade 3 SerCa; right tube intramucosal carcinoma; bilateral ovaries grade 3 SerCa NA Unknown Yes No 
    27 57 Right tube grade 3 SerCa; bilateral surface ovarian grade 3 SerCa NA Unknown No No 
    28 59 Right tube grade 3 SerCa, metastasis to left tubal serosa and ovarian surface; right ovary NDH NA Unknown No No 
    29 57 Right tube grade 3 SerCa, metastasis to right ovary; left ovary primary SerCa NA Unknown Yes No 
    30 58 Left tube grade 3 SerCa; right tube mucosal epithelial hyperplasia; bilateral ovaries grade 3 SerCa NA BRCA1 Yes Yes 
Ovarian SerCa       
    31 54 Bilateral ovaries grade 3 transitional cell carcinoma with focal serous differentiation NA BRCA1 Yes Yes 
    32 55 Ovarian grade 3 papillary SerCa NA Unknown Yes No 
    33 50 Bilateral ovarian grade 3 papillary SerCa, extension through capsule into right fallopian tube; left tube NDH NA BRCA1 917delTT Yes No 
    34 42 Bilateral ovaries and tubes grade 3 SerCa NA BRCA1 3875delGTCT No Yes 
    35* 57 See 26 NA Unknown Yes No 
    36 57 See 27 NA Unknown No No 
    37 32 Bilateral ovaries and tubes grade 3 SerCa NA BRCA1 Yes No 
SampleAgeIndication for surgery/diagnosisOvarian cycle statusBRCA1/2 statusFamily historyPrevious breast cancer
FTEn       
    1 46 History of cervical carcinoma Luteal NA No No 
    2 46 Myometrial leiomyoma Luteal NA No No 
    3 42 Grade 3 invasive squamous cell carcinoma of cervix; adenomyosis Luteal NA No No 
    4 36 Adenocarcinoma in situ of endocervix Luteal NA No No 
    5 43 Left ovary serous cystadenoma; adenomyosis Luteal NA No No 
    6 47 Uterus leiomyoma Luteal NA No No 
    7 41 Retriperitoneal high grade leiomyosarcoma Follicular NA No No 
    8 42 Grade 1 invasive adenocarcinoma of cervix; adenomyosis Follicular NA No No 
    9 46 NDH Follicular NA No No 
    10 47 History of cervical carcinoma Follicular NA No No 
    11 50 History of cervical carcinoma Follicular NA No No 
    12 48 Myometrial and subserosal uterine leiomyoma Follicular NA No No 
FTEb       
    13 44 Bilateral ovaries multiple follicular cysts; leiomyomata Luteal BRCA1 5382insC Yes No 
    14 45 Myometrial leiomyoma Luteal BRCA1 185delAG Yes No 
    15 38 NDH Luteal BRCA1 A1959T Yes No 
    16 40 Bilateral ovarian endometriosis; myometrium leiomyoma Luteal BRCA1 185delAG Unknown No 
    17 39 NDH Luteal BRCA1 185delAG Yes No 
    18 46 Right ovary cortical epithelial inclusions with hyperplasia and serous tubal metaplasia; left ovary serous cyst Luteal BRCA2 C5910G Yes No 
    19 42 Myometrium leiomyoma Follicular BRCA1 5382insC Yes Yes 
    20 40 Right ovary mature cystic teratoma negative for malignancy Follicular BRCA1 185delAG Yes No 
    21 47 Leiomyomata Follicular BRCA2 6174delT Yes No 
    22 39 Bilateral ovaries cystic follicles negative for malignancy Follicular BRCA1 1293del40 Yes No 
    23 37 NDH Follicular BRCA1 185delAG Unknown Yes 
    24 39 NDH Follicular BRCA1 185delAG Yes Yes 
Fallopian tube SerCa       
    25 41 Right tube poorly differentiated carcinoma favor serous type, grade 3; bilateral ovaries negative for malignancy NA Negative No No 
    26* 57 Left tube grade 3 SerCa; right tube intramucosal carcinoma; bilateral ovaries grade 3 SerCa NA Unknown Yes No 
    27 57 Right tube grade 3 SerCa; bilateral surface ovarian grade 3 SerCa NA Unknown No No 
    28 59 Right tube grade 3 SerCa, metastasis to left tubal serosa and ovarian surface; right ovary NDH NA Unknown No No 
    29 57 Right tube grade 3 SerCa, metastasis to right ovary; left ovary primary SerCa NA Unknown Yes No 
    30 58 Left tube grade 3 SerCa; right tube mucosal epithelial hyperplasia; bilateral ovaries grade 3 SerCa NA BRCA1 Yes Yes 
Ovarian SerCa       
    31 54 Bilateral ovaries grade 3 transitional cell carcinoma with focal serous differentiation NA BRCA1 Yes Yes 
    32 55 Ovarian grade 3 papillary SerCa NA Unknown Yes No 
    33 50 Bilateral ovarian grade 3 papillary SerCa, extension through capsule into right fallopian tube; left tube NDH NA BRCA1 917delTT Yes No 
    34 42 Bilateral ovaries and tubes grade 3 SerCa NA BRCA1 3875delGTCT No Yes 
    35* 57 See 26 NA Unknown Yes No 
    36 57 See 27 NA Unknown No No 
    37 32 Bilateral ovaries and tubes grade 3 SerCa NA BRCA1 Yes No 

Abbreviations: NDH, no diagnostic histopathology observed at pathologic review; NA, data points that are not applicable/not evaluated at the time of case selection.

*

Two independent carcinoma samples obtained from the same patient.

Two independent carcinoma samples obtained from same patient.

Laser capture microdissection and RNA extraction. To eliminate potential bias and minimize inclusion of stromal cells in all profiled cases, materials for gene expression analysis of FTEn, FTEb, and SerCa specimens were obtained using the same protocol of laser capture microdissection followed by linear amplification. Shortly before laser capture microdissection, 7-μm frozen sections were cut, adhered onto Superfrost slides (Fisher Scientific Company), and immediately stored on dry ice. Additional 5-μm sections were cut at the beginning and end of each sample and stained with H&E for review to ensure proper tissue orientation and histology. Epithelial cells were then obtained from all specimens using the Arcturus PixCell IIe Laser Capture Microdissection system (Arcturus) following manufacturer's instructions. Collected cells were immediately stored at −80°C before extraction of total RNA using a Stratagene Absolutely RNA MicroPrep kit (Stratagene). Sufficient quality and quantity of RNA was confirmed using an Agilent 2100 Bioanalyzer RNA 6000 Pico LabChip kit (Agilent Technologies, Inc.) and NanoDrop ND-1000 Spectrophotometer (NanoDrop Technologies), respectively, before official inclusion in the study.

Linear amplification and hybridization to GeneChip arrays. Ten nanograms of total RNA from study cases were reverse transcribed and linearly amplified using the NuGEN Biotin Ovation kit (NuGEN Technologies) in collaboration with the University Health Network Microarray Center. This method has previously been shown to provide sufficient material for global gene expression analysis from limited total RNA, while accurately and reproducibly maintaining differential transcript abundance in nonamplified samples (21). Briefly, first-strand cDNA synthesis was done using a unique DNA/RNA primer, followed by second strand cDNA synthesis and SPIA isothermal linear amplification. Amplified cDNA was then purified, enzymatically fragmented, and labeled with biotin. Quality and quantity of the purified labeled cDNA product were confirmed before hybridization to Affymetrix GeneChip U133A Plus 2.0 arrays (Affymetrix, Inc.) according to manufacturer's instructions.7

7

Protocols available at http://www.microarrays.ca/.

Amplification and hybridization of samples was done in six separate runs, with each group represented in each run (FTEn from luteal and follicular phase, FTEb from luteal and follicular phase and tubal and ovarian SerCa). Profiling data for all samples has been deposited in the National Center for Biotechnology Information Gene Expression Omnibus8 and are accessible through Gene Expression Omnibus Series accession number GSE10971.

Statistical analysis of gene expression profiles. All CEL files were imported into ArrayAssist (Stratagene), and a master table of all hybridizations was created using the robust multiarray average algorithm. Acceptable array performance was confirmed by review of generated RPT files. Robust multiarray average normalized data were then used as the basis for all subsequent clustering and statistical analysis. Unsupervised binary tree-structured vector quantization clustering was done using all SerCa samples. This method combines tree-structured vector quantization and partitive k-means clustering to separate samples into homogenous groups based on similar levels of gene expression (22). ArrayAssist was used to perform unsupervised hierarchical clustering of all SerCa cases following definition of tubal SerCa as baseline. Hierarchical clustering was subsequently done on all hybridizations following definition of FTEn samples as baseline, as well as on data grouped by major class (FTEn, FTEb, or SerCa) or subgroup (FTEn luteal, FTEn follicular, FTEb luteal, FTEb follicular, fallopian tube SerCa, ovarian SerCa) after defining FTEn and FTEn luteal as baseline, respectively.

Robust multiarray average normalized data were then imported into Microsoft Excel for identification of differentially expressed genes using significance analysis of microarrays (SAM version 3.0).9

For each comparison, two-class unpaired analysis was done using unlogged data, and the δ value was selected based on an acceptable corresponding median false discovery rate (FDR). To find overlapping significant probe sets, relevant significance lists were imported into ArrayAssist and compared using the Venn diagram function. Additional information for probe sets of interest, including gene ontology, was obtained using the batch query function on the NetAffx Analysis Center portion of the Affymetrix Web site.10

Identification of candidate genes using protein-protein interaction networks. To visualize the interactive networks of genes found significantly altered, as well as to aid in the selection of the most promising candidates for follow-up studies, a combination of bioinformatic tools was used. Lists of significant probe set IDs separated by analysis and direction of change in the group of interest were submitted to the I2D (Interlogous Interaction Database) Web site11

(version 1.6) to obtain first-degree interaction information for the corresponding proteins (23). Next, known and predicted protein-protein interactions for each list were visualized using NAViGaTOR (Network Analysis, Visualization, and Graphing TORonto) software, version 1.1.0, to elucidate the connectivity of the significantly altered genes.12 The extent of overlap of individual networks of interest was visualized using the Compare Networks (union) option. Proteins of extreme interest (p53 and BRCA1) were then located by Swiss Prot ID, and all noninteracting proteins were deleted.

Real-time quantitative reverse transcription–PCR. Three representative samples from each nonmalignant group (FTEn luteal, FTEn follicular, FTEb luteal, FTEb follicular) and six SerCa samples were selected for validation of gene expression profiles. Ten nanograms of RNA were linear amplified using the NuGEN Ovation RNA Amplification System (NuGEN Technologies). RNA was also isolated from SKOV3 ovarian cancer cells using TRIzol reagent (Invitrogen Corporation) for use as a calibrator sample. Contaminating DNA was removed (DNA-free kit, Ambion, Inc.), and first-strand cDNA was generated using SuperScript III Reverse Transcriptase and Oligo(dT)20 primers (Invitrogen). Quality and quantity of all amplified cDNA were confirmed, and all samples were diluted to 1.6 ng/μL using sterile double distilled water.

Primer Express Software version 2.0 (Applied Biosystems) was used to select specific primers for human disabled homolog 2 mitogen-responsive phosphoprotein (DAB2), Ski-like (SKIL), and reference gene β-actin (ACTB) based on the sequence of the relevant probe set(s). All amplicons were situated within the first 1,500 bp from the poly(A) tail, given the use of oligo(dT)-primed reverse transcription of study and calibrator samples. Selected primer sequences included DAB2 forward 5′-CTAGCTATTGCAAATGAGGGAAG-3′, DAB2 reverse 5′-GGTAATACTACTTGAACCCAGGAGCA-3′, SKIL forward 5′AAAACTGTCCTCTACCTCACGTG-3′, SKIL reverse 5′-AAGCTCAGAGCACAGTATGTCATG-3′, ACTB forward 5′-GCATTGTTACAGGAAGTCCCTTG-3′, and ACTB reverse 5′-CTATCACCTCCCCTGTGTGGA-3′. End-point PCR was done for all primer sets, and the reaction product was sequence verified.

Real-time quantitative PCR was done using the QuantiTect SYBR Green PCR kit (Qiagen, Inc.) according to manufacturer's instructions. PCR was done using the ABI PRISM 7900HT Sequence Detection System (Applied Biosystems), with use of the ROX internal reference dye. A dissociation reaction was done at the end of the PCR program to confirm amplification of a single product at the expected melting temperature, and the product was run on a 2.5% agarose gel to confirm correct size. All experiments included triplicate wells of each sample for both target and reference gene. The comparative CT method for relative quantitation was done using ABI PRISM Sequence Detection Software version 2.0 (Applied Biosystems). Target gene CT values were normalized to ACTB. Statistical analysis was done using one-way ANOVA followed by a least significant difference test for post hoc comparisons (P < 0.05).

Immunohistochemistry. A tissue microarray consisting of a larger subset of nonmalignant fallopian tube specimens from control patients and mutation carriers was constructed using the semiautomated TMArrayer (Pathology Devices, Inc.). Patient samples were identified according to history and diagnosis from the University Health Network Ovarian Tissue Bank and Database. Routine H&E sections were examined and a 1.5 mm2 area of histologically normal FTE was selected before transfer of the corresponding formalin-fixed paraffin-embedded tissue core to the master block. The final array consisted of a total of 33 nonmalignant specimens from confirmed BRCA1/2 mutation carriers (including 11 luteal, 16 follicular, 3 early follicular, and 3 perimenopausal), 25 normal control specimens (including 11 luteal, 12 follicular, and 2 perimenopausal), and 2 cores each of normal kidney and liver for orientation purposes. All nonmalignant FTEn and FTEb cases used for gene expression profiling were included. Duplicate 5 μm sections of the array were included on each slide to control for potential variability during immunohistochemistry. In addition, a separate array containing duplicate 0.6 mm2 cores obtained from 59 cases of high-grade SerCa was also used to assess the expression of candidates in both hereditary and sporadic carcinoma tissue specimens (average age, 61.8 y; 95% CI, 59.1-64.5). All carcinoma specimens were obtained from the Ovarian Tissue Bank and Database and were naive to chemotherapy.

Immunohistochemistry was done using standard procedures following microwave antigen retrieval using goat polyclonal anti-SKIL antibody [SnoN (K-20), Santa Cruz Biotechnology] at a dilution of 1 in 600, mouse monoclonal anti-DAB2 (disabled-2/p96 antibody, BD Biosciences) at a dilution of 1 in 50, or mouse monoclonal anti-cdc2 [cdc2 (P0H1), Cell Signaling Technology, Inc.] at a dilution of 1 in 50. Antibody concentrations were optimized on normal or malignant breast tissue sections before staining tissue array slides.

After immunohistochemistry, the ScanScope CS slide scanner (Aperio Technologies, Inc.) was used to create digital slide images at 20× magnification. Tissue cores were then visualized and manually scored using ImageScope software (Aperio Techologies), version 6.25. Scoring was independently done by two observers (A.A.T. and P.A.S.) blinded to patient information. Scores were given for percentage of epithelial cells stained (0, 0%; 1, 1-24%; 2, 25-50%; 3, >50%), as well as intensity of staining (0, negative; 1, light; 2, medium; 3, dark). The combined histoscore was obtained by adding these two individual scores, and nuclear and cytoplasmic staining was reviewed independently (24). Statistical analysis was done using ANOVA followed by least significant difference (P < 0.05). Comparison of unpaired proportions was by Fisher's exact test (P < 0.05).

High-grade ovarian SerCa and fallopian tube SerCa exhibit indistinguishable gene expression profiles. An initial critical question was whether SerCa designated as of ovarian origin, according to current pathology practice, is molecularly similar to that apparently derived from the fallopian tube. A high degree of similarity would support the idea that the FTE is the cell of origin for both tubal and ovarian SerCa. Unsupervised hierarchical (agglomerative) clustering of the individual SerCa samples after robust multiarray average normalization showed that the tubal and ovarian specimens had closely related global gene expression profiles (Fig. 1A). SerCa of presumed ovarian origin clustered together, with many of the tubal cancers clustering more closely with the ovarian cancers than with other tubal cancers. To examine this relationship more closely, an unsupervised partitive clustering method, binary tree-structured vector quantization, was used. This method iteratively partitioned the 13 cases by a k-means algorithm (k=2), considering the partitioning of all probe set responses by a self-organizing map algorithm. This method clearly revealed that samples partitioned together irrespective of presumed origin or known mutation status (Fig. 1B). Although similar, identical results were not obtained using the two clustering methods. This is likely due to the distinct approaches (agglomerative versus partitive), as well as the differential sensitivity of each method to data normalization (22). Two-class paired SAM analysis of the two tubal and ovarian cancers from the same patients revealed no differentially expressed genes at a minimum FDR of 40%, as did two-class unpaired analysis of the remaining carcinoma specimens at an FDR of 21%. Thus, cluster analysis, as well as SAM, indicates that SerCas have similar molecular profiles whether of presumed ovarian or tubal origin.

Fig. 1.

Comparison of fallopian tube and ovarian SerCas. Unsupervised hierarchical (agglomerative, A) and binary tree-structured vector quantization (partitive, B) clustering was done using all grade 3 SerCas, revealing the similarity of samples of presumed tubal and ovarian origin. Numbers shown at terminal ends in A and B represent sample numbers, as indicated in Table 1. A, FT, fallopian tube carcinomas; Ov, ovarian carcinomas. B, ovarian and tubal cancers associated with a known BRCA1/2 mutation and/or family history are represented by open and closed bars, respectively. Ovarian and tubal cancers with unknown familial status are represented by hatched and spotted bars, respectively. C, Venn diagrams were used to compare probe sets differentially expressed by SAM between FTE from normal controls and tubal and ovarian carcinomas. At an FDR of 4.6%, 5,522 of 11,574 of the total probe sets (48%) were found to be altered in tumors of both tubal and ovarian origin (boxed).

Fig. 1.

Comparison of fallopian tube and ovarian SerCas. Unsupervised hierarchical (agglomerative, A) and binary tree-structured vector quantization (partitive, B) clustering was done using all grade 3 SerCas, revealing the similarity of samples of presumed tubal and ovarian origin. Numbers shown at terminal ends in A and B represent sample numbers, as indicated in Table 1. A, FT, fallopian tube carcinomas; Ov, ovarian carcinomas. B, ovarian and tubal cancers associated with a known BRCA1/2 mutation and/or family history are represented by open and closed bars, respectively. Ovarian and tubal cancers with unknown familial status are represented by hatched and spotted bars, respectively. C, Venn diagrams were used to compare probe sets differentially expressed by SAM between FTE from normal controls and tubal and ovarian carcinomas. At an FDR of 4.6%, 5,522 of 11,574 of the total probe sets (48%) were found to be altered in tumors of both tubal and ovarian origin (boxed).

Close modal

In contrast, comparison of all FTEn samples with all cancers revealed 4,263 probe sets with decreased expression and 7,311 probe sets with increased expression in the cancers at an FDR of 4.3%. Of the down-regulated probe sets, 2,235 (52%) were shared between the two malignancies, as were 3,287 (45%) of the up-regulated probe sets (Fig. 1C). Given the high degree of similarity between fallopian tube SerCa and ovarian SerCas, all subsequent comparisons to malignant samples were done combining the SerCas as a single group.

Differential expression of genes in a subset of histologically normal FTEb samples. Hierarchical clustering using all probe sets was done to determine the overall similarity of FTEb and SerCa samples. Analysis using grouped data revealed that FTEb samples, which were histologically indistinguishable from FTEn, clustered with SerCa samples rather than normal controls (Fig. 2A), demonstrating that gene expression altered in mutation carriers is consistent with changes that have occurred in malignant cells. These changes likely reflect and contribute to the overall increased risk for malignant transformation in mutation carriers, consistent with the documented roles of BRCA1 and BRCA2 proteins in maintaining genomic integrity through participation in DNA repair, transcriptional regulation, and cell cycle control (2527).

Fig. 2.

Identification of differentially expressed genes in histologically normal FTEb. Hierarchical clustering was done using grouped data (A), revealing the overall similarity of FTEb and SerCa specimens. Unsupervised clustering was then done using individual hybridizations (B) to identify specific FTEb samples more closely resembling SerCa specimens compared with normal controls (boxed). Numbers shown at terminal ends in A and B represent sample numbers as indicated in Table 1. B, FTEn samples obtained during the luteal and follicular phase (pink and light blue, respectively), FTEb luteal and follicular samples (red and dark blue, respectively), and tubal and ovarian SerCa samples (green and purple, respectively). SAM revealed the decreased expression of 288 probe sets and increased expression of 598 probe sets between these samples and the remaining FTEb specimen grouping with normal controls at 4.3% FDR [average fold-change of 2.9 (95% CI, 2.5-3.2) for decreased probe sets and 3.7 (95% CI, 3.4-4.0) for increased]. The distribution of primary ontologies of the genes represented by these probe sets (C). Venn diagrams were used to compare significantly altered probe sets to those differentially expressed between SerCa and FTEn specimens at the same FDR (D). Overlapping genes (boxed).

Fig. 2.

Identification of differentially expressed genes in histologically normal FTEb. Hierarchical clustering was done using grouped data (A), revealing the overall similarity of FTEb and SerCa specimens. Unsupervised clustering was then done using individual hybridizations (B) to identify specific FTEb samples more closely resembling SerCa specimens compared with normal controls (boxed). Numbers shown at terminal ends in A and B represent sample numbers as indicated in Table 1. B, FTEn samples obtained during the luteal and follicular phase (pink and light blue, respectively), FTEb luteal and follicular samples (red and dark blue, respectively), and tubal and ovarian SerCa samples (green and purple, respectively). SAM revealed the decreased expression of 288 probe sets and increased expression of 598 probe sets between these samples and the remaining FTEb specimen grouping with normal controls at 4.3% FDR [average fold-change of 2.9 (95% CI, 2.5-3.2) for decreased probe sets and 3.7 (95% CI, 3.4-4.0) for increased]. The distribution of primary ontologies of the genes represented by these probe sets (C). Venn diagrams were used to compare significantly altered probe sets to those differentially expressed between SerCa and FTEn specimens at the same FDR (D). Overlapping genes (boxed).

Close modal

Unsupervised clustering using individual data revealed that four BRCA1-mutated FTEb samples clustered with all of the carcinomas, indicating that global gene expression in this subset of samples most closely resembles that of malignant epithelial cells (Fig. 2B). Whereas it is not possible to determine if these cases would have been more likely to progress to SerCa had prophylactic surgery not occurred, it is feasible that these samples have acquired gene expression changes involved in serous carcinogenesis that have not yet resulted in atypical histology. SAM two-class unpaired analysis identified 288 probe sets with decreased expression and 598 with increased expression in these four samples relative to the remaining FTEb samples that were more similar to normal controls at an FDR of 4.3%. Gene ontology analysis revealed that many differentially expressed probe sets correspond to genes with known roles in BRCA1/2-dependent processes (transcriptional regulation, cell cycle control, ubiquitin cycle; refs. 25, 27), as well as others involved in tumor initiation and progression in general (apoptosis, cell adhesion, and cell motility; Fig. 2C; see Supplementary Table S1 for a complete list of significantly altered genes arranged by gene ontology). In addition, 181 of 288 (63%) of the probe sets with decreased expression and 172 of 598 (29%) with increased expression overlapped in the same direction as those differentially expressed between nonmalignant FTEn and SerCa samples as a group at the same FDR (4.3%), further supporting the idea that these may include some of the earliest events in serous carcinogenesis (Fig. 2D).

Differential effect of the ovarian cycle in BRCA1/2 mutation carriers. Given the unexpected finding that all four FTEb samples grouping with SerCa were obtained during the luteal phase, we further explored the association of the ovarian cycle with FTE gene expression. Hierarchical clustering of all samples using all probe sets was repeated using data grouped by both BRCA mutation and ovarian cycle status. The dendrogram obtained confirmed that FTEb samples from the luteal phase (FTEb luteal) more closely resembled SerCa samples compared with FTEb samples from the follicular phase (FTEb follicular; Fig. 3A).

Fig. 3.

The effect of the ovarian cycle on FTE gene expression in BRCA1/2 mutation carriers and normal controls. Hierarchical clustering using data grouped by mutation status and stage of the ovarian cycle at the time of surgery (A) confirmed the similarity of FTEb luteal and SerCa samples. SAM identified a greatly increased number of differentially expressed probe sets by ovarian cycle in mutation carriers compared with normal controls at an FDR of 9.8% (B). All differentially expressed probe sets in the normal controls were increased in the follicular phase, at an average fold-change of 2.7 (95% CI, 2.2-3.3) compared with luteal samples. Of the 392 differentially expressed probe sets in mutation carriers, 322 were increased in the follicular phase at an average fold-change of 2.7 (95% CI, 2.4-2.9) and 70 were increased in the luteal phase at an average fold-change of 3.4 (95% CI, 2.9-3.9) relative to follicular samples. The distribution of primary ontologies of the genes represented by the probe sets differentially expressed in mutation carriers but not normal controls (C). Venn diagrams were used to compare probe sets significantly altered in FTEb luteal samples to those differentially expressed between SerCa and FTEn specimens at an FDR of 8.3% (D).

Fig. 3.

The effect of the ovarian cycle on FTE gene expression in BRCA1/2 mutation carriers and normal controls. Hierarchical clustering using data grouped by mutation status and stage of the ovarian cycle at the time of surgery (A) confirmed the similarity of FTEb luteal and SerCa samples. SAM identified a greatly increased number of differentially expressed probe sets by ovarian cycle in mutation carriers compared with normal controls at an FDR of 9.8% (B). All differentially expressed probe sets in the normal controls were increased in the follicular phase, at an average fold-change of 2.7 (95% CI, 2.2-3.3) compared with luteal samples. Of the 392 differentially expressed probe sets in mutation carriers, 322 were increased in the follicular phase at an average fold-change of 2.7 (95% CI, 2.4-2.9) and 70 were increased in the luteal phase at an average fold-change of 3.4 (95% CI, 2.9-3.9) relative to follicular samples. The distribution of primary ontologies of the genes represented by the probe sets differentially expressed in mutation carriers but not normal controls (C). Venn diagrams were used to compare probe sets significantly altered in FTEb luteal samples to those differentially expressed between SerCa and FTEn specimens at an FDR of 8.3% (D).

Close modal

Ovarian steroids are known to affect the growth and differentiation of the FTE. Estrogens act during the follicular phase to promote ciliogenesis and secretory cell hypertrophy, whereas progesterone, which is predominant during the luteal phase, induces dedifferentiation (28, 29). However, these hormonal influences are not widespread within the tube and can be quite variable among individuals; thus, both ciliated and secretory cells are present in the FTE throughout the cycle. Interestingly, SAM two-class unpaired analysis revealed that the number of differentially expressed probe sets between the luteal and follicular phases was far greater in mutation carriers than in normal controls. At an FDR of 9.8%, only 21 probe sets were found to be decreased in the luteal relative to the follicular phase in normal control patients (Fig. 3B). In contrast, 322 probe sets were decreased and 70 were increased in the luteal relative to follicular phase in FTEb at the same FDR, suggesting that hormonal influences on the FTE may be altered as a result of reduced functional BRCA levels. Accordingly, the average BRCA1 level in FTEb luteal samples was 50% lower than that seen in FTEb follicular samples, and an 80% decrease was observed in the four FTEb luteal samples clustering with SerCa relative to the remaining FTEb specimens (not shown). Previous studies have implicated both BRCA1 and BRCA2 in the regulation of gonadal steroid hormone receptor activity (30, 31), and this regulation may contribute to the differences in gene expression between control patients and BRCA1/2 mutation carriers.

The 389 probe sets specific to mutation carriers were subjected to gene ontology analysis and were found to correspond to BRCA1/2-dependent and tumorigenesis-involved processes similar to that observed in the previous analysis (Fig. 3C; see Supplementary Table S2 for a complete list of significantly altered genes arranged by gene ontology). Interestingly, 114 of 319 of the probe sets with decreased expression (36%) and 26 of 70 with increased expression (37%) in FTEb luteal samples overlapped in the same direction with probe sets differentially expressed between FTEn and SerCa samples at a similar FDR (8.3%; Fig. 3D). This is in contrast to an overlap of 9 of 70 with decreased expression (13%; P = 0.0001) and 59 of 319 with increased expression (18%; P = 0.001) in FTEb follicular samples. That the profiles obtained during the luteal phase of mutation carriers most closely resembles the gene expression pattern of SerCa raises the possibility that the hormonal milieu associated with the luteal phase may contribute to cancer predisposition in some individuals, such as BRCA1/2 mutation carriers.

A primary hormonal difference between the follicular and luteal phases is the elevation of circulating progesterone during the luteal phase. Studies have frequently suggested that progesterone is a protective rather than exacerbating factor in ovarian cancer development (32, 33). Epidemiologic data generally show that women with conditions in which progesterone is elevated have a lower incidence of ovarian cancer. For instance, the protective effect of increased parity and twin pregnancies has been attributed to the greatly increased level of maternal circulating progesterone (17, 18, 32, 34). In addition, many studies have linked the use of combination oral contraceptives to a decreased ovarian cancer risk in mutation carriers and the general population. A few studies have further suggested that formulations with high progestin potency offer greater protection than those with low progestin potency (30, 32, 35, 36). Finally, treatment of normal ovarian surface epithelium and ovarian cancer cells with progesterone has a growth inhibitory effect (32, 34). Accordingly, some authors have suggested that exposure to progesterone may eliminate cells genetically damaged by incessant ovulatory events through induction of apoptosis, thus providing an exfoliation effect (32, 34). Evidence has emerged, however, that the effects of progesterone may be more complex than previously appreciated. For instance, whereas high concentrations of progesterone suppressed tumorigenesis in nude mice inoculated with ovarian cancer cells (37), lower concentrations increased the proliferative capacity of ovarian surface epithelium and ovarian cancer cells in culture (38). This is consistent with the postulate that luteal-phase levels of progesterone have a growth promotional effect, whereas the higher levels achieved during pregnancy or oral contraceptive use induce cell cycle arrest or apoptosis of epithelial cells (32). Similar studies have not yet been reported for FTE cells. However, it is possible that exposure to luteal-phase levels of progesterone may not be protective against malignant transformation of the epithelial cells of the distal fallopian tube, similar to the effect observed in the breast (3941), particularly in cells in which the DNA repair pathways may be compromised. It is of course possible that other hormonal changes associated with the luteal phase may affect gene expression in the FTE, and further studies are required using FTE cells in culture. Finally, while all nonmalignant cases in this study have been age-matched, there is evidence that mutation carriers initiate menopause at an earlier age than normal controls (42), suggesting that the nature of the hormonal milieu of the luteal phase may be different in the two populations.

Identification of protein-protein interaction networks potentially involved in initiation of SerCa. To select genes for validation and further investigation, we queried the I2D protein interaction database with our set of differentially expressed genes from both SAM analyses. Of the 886 probe sets differentially expressed in the four FTEb sample grouping with SerCa compared with the remaining FTEb, 417 (47%) mapped to I2D, as well as 218 of 389 (56%) of the probe sets differentially expressed in FTEb luteal compared with FTEb follicular samples. Because of the known involvement of p53 in high-grade SerCa, we focused our attention on differentially expressed genes encoding for proteins predicted to directly or indirectly interact with BRCA1/2 or p53 proteins. Because mutation of BRCA1 or BRCA2 leads to activation of the damage response pathway, loss of this pathway by somatic mutation of p53 and presumably other members is required for malignant transformation (43). Consequently, accumulation of p53 protein is frequently observed in the histologically normal FTE of mutation carriers and control patients, and decreased expression of cell cycle arrest genes p21 and p27 have been observed in prophylactic tubal specimens relative to normal controls (8, 13). Supplementary Figs. S1 and S2 show the networks obtained through I2D visualized with NAViGaTOR software. One gene found in networks of both comparisons, DAB2, was found to be decreased in FTEb luteal and SerCa samples. DAB2 is an adaptor molecule that exerts its tumor suppressive function largely through its role in facilitating transforming growth factor-β (TGF-β)–induced growth inhibition (44), a pathway that must be circumvented in early stages of epithelial carcinogenesis and which has previously been shown to be dysfunctional in ovarian cancer (45). In addition, we selected SKIL, which was up-regulated in FTEb luteal samples and acts to inhibit Smad2 and Smad3 (46), targets that are enhanced by DAB2 (Fig. 4). The implication of expression changes in the TGF-β pathway regulatory genes DAB2 and SKIL in FTEb samples during the luteal phase is highlighted by the observation that 15% of the significantly altered probe sets found by both SAM analyses (185 of 1,201 unique probe sets) correspond to genes known to be downstream TGF-β targets (4750), consistent with an altered TGF-β response (see Supplementary Tables S1 and S2). Such genes include DAB2 (47, 49) and SKIL (48, 49) themselves, in addition to well-established TGF-β targets ID2, ETS2, and PIM1 (50).

Fig. 4.

Protein-protein interaction subnetwork potentially involved in initiation of SerCa. Submission of probe sets differentially expressed in the FTEb specimens that clustered with SerCa, as well as those specifically altered in FTEb luteal samples to the online Interlogous Interaction Database (I2D, version 1.6), revealed overlapping networks of proteins with altered expression in the four FTEb samples and/or FTEb luteal samples overall (full networks shown in Supplementary Figs. S1 and S2). The interesting subnetwork containing the TGF-β pathway regulators DAB2 and SKIL is expanded here. Genes encoding proteins shown as upward-pointing triangles were increased in FTEb luteal samples, whereas those shown as downward-pointing triangles were decreased in the same samples. Known and predicted interactions between proteins are indicated by a line, with activation represented by an arrowhead and inhibition represented by a blunt end. *, TGF-β target genes identified by select gene expression profiling studies (4750).

Fig. 4.

Protein-protein interaction subnetwork potentially involved in initiation of SerCa. Submission of probe sets differentially expressed in the FTEb specimens that clustered with SerCa, as well as those specifically altered in FTEb luteal samples to the online Interlogous Interaction Database (I2D, version 1.6), revealed overlapping networks of proteins with altered expression in the four FTEb samples and/or FTEb luteal samples overall (full networks shown in Supplementary Figs. S1 and S2). The interesting subnetwork containing the TGF-β pathway regulators DAB2 and SKIL is expanded here. Genes encoding proteins shown as upward-pointing triangles were increased in FTEb luteal samples, whereas those shown as downward-pointing triangles were decreased in the same samples. Known and predicted interactions between proteins are indicated by a line, with activation represented by an arrowhead and inhibition represented by a blunt end. *, TGF-β target genes identified by select gene expression profiling studies (4750).

Close modal

Differential expression of SKIL and DAB2 in FTEb samples during the luteal phase. In agreement with our Affymetrix data, FTEb luteal samples were found to express higher levels of SKIL mRNA compared with FTEb follicular samples as determined by real-time quantitative reverse transcription–PCR, although this did not reach statistical significance (Fig. 5A, left). A further increase in SKIL mRNA was observed in SerCa samples compared with FTEb follicular (P < 0.05). Immunohistochemistry on tissue microarrays revealed a similar average intensity of nuclear staining in FTEb luteal and SerCa samples, as well as a trend toward increased staining in FTEb luteal compared with FTEb follicular samples (Fig. 5A, right). Representative images of SKIL immunohistochemical staining are shown in Fig. 5B.

Fig. 5.

Expression of SKIL, DAB2, and cdc2 in FTEb luteal and SerCa samples. To confirm the increased expression of SKIL and decreased expression of DAB2 in FTEb luteal and SerCa samples, real-time quantitative reverse transcription–PCR was done using cDNA generated from a subset of samples previously used for gene expression profiling (n = 3 for each nonmalignant group, as well as six SerCa). Immunohistochemistry was also done on tissue microarrays containing a total of 58 nonmalignant FTE from BRCA1/2 mutation carriers and normal controls from both phases of the ovarian cycle, as well as 59 grade 3 SerCas. Real-time quantitative reverse transcription–PCR (A, left) and immunohistochemistry on tissue microarrays (A, right) confirmed a trend toward increased average expression of SKIL in FTEb luteal and SerCa samples compared with FTEb follicular and normal controls (images shown in B). Magnification, 40×. Real-time quantitative reverse transcription–PCR (C, left) and immunohistochemistry (C, right) also confirmed the decreased average expression of DAB2 in FTEb luteal and SerCa samples compared with FTEb follicular specimens (images shown in D). Magnification, 40× (top). Specific retention of DAB2 staining in the secretory (solid arrow) compared with ciliated (dashed arrow) cells of the mucosal lining in FTEb follicular samples is highlighted. This differential pattern of staining was also observed in normal control patients, although the difference between the histoscore of FTEn luteal and FTEn follicular samples did not attain statistical significance. Similar to SKIL and in contrast to DAB2, cdc2 protein showed increased nuclear intensity in FTEb luteal (n = 11) and SerCa (n = 51) samples compared with FTEb follicular (n = 16) specimens (images shown in D). Magnification, 40× (bottom). FTEb luteal samples had an increased average nuclear histoscore compared with FTEb follicular samples, although this did not attain statistical significance (4.91; 95% CI, 4.50-5.32 versus 4.20; 95% CI, 3.72-4.68). No difference in staining was observed in luteal (n = 11) and follicular (n = 12) samples from normal controls (data not shown). Vertical bars shown in A and C represent average mRNA or protein expression for each group of samples, with statistically significant differences in average mRNA or protein expression indicated by different letters (one-way ANOVA with least significant difference post hoc test; P < 0.05). Black circles in A and C indicate the individual sample values for real-time quantitative reverse transcription–PCR.

Fig. 5.

Expression of SKIL, DAB2, and cdc2 in FTEb luteal and SerCa samples. To confirm the increased expression of SKIL and decreased expression of DAB2 in FTEb luteal and SerCa samples, real-time quantitative reverse transcription–PCR was done using cDNA generated from a subset of samples previously used for gene expression profiling (n = 3 for each nonmalignant group, as well as six SerCa). Immunohistochemistry was also done on tissue microarrays containing a total of 58 nonmalignant FTE from BRCA1/2 mutation carriers and normal controls from both phases of the ovarian cycle, as well as 59 grade 3 SerCas. Real-time quantitative reverse transcription–PCR (A, left) and immunohistochemistry on tissue microarrays (A, right) confirmed a trend toward increased average expression of SKIL in FTEb luteal and SerCa samples compared with FTEb follicular and normal controls (images shown in B). Magnification, 40×. Real-time quantitative reverse transcription–PCR (C, left) and immunohistochemistry (C, right) also confirmed the decreased average expression of DAB2 in FTEb luteal and SerCa samples compared with FTEb follicular specimens (images shown in D). Magnification, 40× (top). Specific retention of DAB2 staining in the secretory (solid arrow) compared with ciliated (dashed arrow) cells of the mucosal lining in FTEb follicular samples is highlighted. This differential pattern of staining was also observed in normal control patients, although the difference between the histoscore of FTEn luteal and FTEn follicular samples did not attain statistical significance. Similar to SKIL and in contrast to DAB2, cdc2 protein showed increased nuclear intensity in FTEb luteal (n = 11) and SerCa (n = 51) samples compared with FTEb follicular (n = 16) specimens (images shown in D). Magnification, 40× (bottom). FTEb luteal samples had an increased average nuclear histoscore compared with FTEb follicular samples, although this did not attain statistical significance (4.91; 95% CI, 4.50-5.32 versus 4.20; 95% CI, 3.72-4.68). No difference in staining was observed in luteal (n = 11) and follicular (n = 12) samples from normal controls (data not shown). Vertical bars shown in A and C represent average mRNA or protein expression for each group of samples, with statistically significant differences in average mRNA or protein expression indicated by different letters (one-way ANOVA with least significant difference post hoc test; P < 0.05). Black circles in A and C indicate the individual sample values for real-time quantitative reverse transcription–PCR.

Close modal

Also consistent with our profiling data, FTEb luteal samples expressed a level of DAB2 mRNA similar to SerCa samples and a greatly reduced expression compared with FTEb follicular samples (P < 0.05; Fig. 5C, left). Immunohistochemistry for DAB2 on tissue microarrays revealed that FTEb luteal and SerCa samples express a decreased level of cytoplasmic DAB2 protein compared with FTEb follicular samples (P < 0.05; Fig. 5C, right). A lower proportion of FTEb luteal (one of nine, 11%) and SerCa (1 of 48, 2%) samples had a histoscore of >4 compared with FTEb follicular specimens (13 of 16, 81%). The decrease in DAB2 during the luteal phase could partly be explained by altered cdc2 protein levels. Previous studies have indicated that functional BRCA1 indirectly inhibits cdc2 kinase activity (51), which then negatively regulates DAB2 through cell cycle–specific phosphorylation (52). Absence of functional BRCA proteins would therefore lead to accumulation of active cdc2 and subsequent reduction of DAB2. Accordingly, in direct opposition to DAB2, there was a trend toward an increased nuclear histoscore for cdc2 in FTEb luteal compared with FTEb follicular samples, and a similar proportion of FTEb luteal (4 of 11, 36%) and SerCa (18 of 51, 35%) samples showed intense nuclear staining. Representative images of DAB2 and cdc2 immunohistochemical staining are shown in Fig. 5D. Most intriguingly, whereas FTEb luteal and SerCa samples showed an overall loss of cytoplasmic DAB2 staining, FTEb follicular samples typically showed retention of staining in the secretory cells of the mucosal lining resulting in an increased overall intensity and histoscore. The specific loss of DAB2 expression in secretory cells during the luteal phase may explain the modest difference in mRNA and protein expression between luteal and follicular samples when including both secretory and ciliated cell populations and further emphasizes the importance of this cell type in serous carcinogenesis (9, 15).

Many studies have observed a similar loss of DAB2 in ovarian carcinomas compared with normal ovarian surface epithelium (5355), although this has not previously been found in the histologically normal ovarian surface epithelium from BRCA1/2 mutation carriers. Interestingly, mice heterozygous for DAB2 frequently develop epithelial dysplasia on the ovarian surface but not malignant ovarian tumors, suggesting that loss of DAB2 is necessary but not sufficient for ovarian carcinoma development (53); however, the oviductal epithelium was not examined. Whereas functional DAB2 exerts its tumor-suppressive effect by mediating TGF-β–induced growth inhibition through transmission of signals from the TGF-β receptors to the Smad transcriptional activators (44), this is opposed by SKIL, which directly represses Smad gene transcription and enhances cytoplasmic sequestration of active Smad protein complexes (46, 56, 57). Although SKIL has not previously been implicated in ovarian cancer development, it lies within a chromosomal region (3q26) previously found to be amplified in a majority of serous fallopian tube and ovarian carcinomas by comparative genomic hybridization (19). SKIL protein is highly elevated in many human cancer cell lines, including ovarian, and has been found to promote tumor growth in nude mice (58). High expression of SKIL protein is also a negative prognostic factor in ER+ breast carcinomas (56). It is therefore likely that the combined effect of decreased DAB2 and increased SKIL in FTEb would promote malignant transformation through potent suppression of TGF-β–induced cell cycle arrest and apoptosis.

In conclusion, gene expression changes potentially involved in the earliest events of tubal and ovarian SerCa have been identified in histologically normal FTE from BRCA1/2 mutation carriers. These expression changes seem to be influenced by reproductive hormones, with components of the luteal phase inducing changes similar to those observed in SerCa specimens. Increased expression of SKIL, coupled with decreased expression of DAB2 in mutation carriers during this phase, could represent some of the earliest initiating or predisposing events of SerCa. Finally, specific loss of DAB2 in the secretory cells of the tubal epithelium during the luteal phase further highlights the relevance of this cell type in SerCa development.

No potential conflicts of interest were disclosed.

Grant support: Toronto Ovarian Cancer Research Network through funds raised by Toronto Fashion Show, Ontario Women's Health Council/Canadian Institute of Health Research Institute of Gender and Health Doctoral Research award (A.A. Tone), Department of Defense Ovarian Cancer Research Program Idea award (P.A. Shaw), and Canadian Institute of Health Research operating grant (T.J. Brown).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

P.A. Shaw and T.J. Brown contributed equally to the work.

We thank Dr. Susan Done and Dr. Igor Jurisica for their critical comments, the University Health Network Biobank for facilitating this research, Kelvin So for his assistance in immunohistochemistry, Neil Winegarden for his guidance in microarray experiments, and the Jurisica group for their support in network analysis of gene expression data.

1
Risch H. Population BRCA1 and BRCA2 mutation frequencies and cancer penetrances: a kin-cohort study in Ontario, Canada.
J Natl Cancer Inst
2006
;
98
:
1694
–705.
2
Aziz S, Kuperstein G, Rosen B, et al. A genetic epidemiological study of carcinoma of the fallopian tube.
Gynecol Oncol
2001
;
80
:
341
–5.
3
Finch A, Beiner M, Lubinski J, et al. Salpingo-oophorectomy and the risk of ovarian, fallopian tube, and peritoneal cancers in women with a BRCA1 or BRCA2 Mutation.
JAMA
2006
;
296
:
185
–92.
4
King MC. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2.
Science
2003
;
302
:
643
–6.
5
Singer G, Kurman RJ, Chang HW, Cho SK, Shih Ie M. Diverse tumorigenic pathways in ovarian serous carcinoma.
Am J Pathol
2002
;
160
:
1223
–8.
6
Barakat RR, Federici MG, Saigo PE, Robson ME, Offit K, Boyd J. Absence of premalignant histologic, molecular, or cell biologic alterations in prophylactic oophorectomy specimens from BRCA1 heterozygotes.
Cancer
2000
;
89
:
383
–90.
7
Casey MJ, Bewtra C, Hoehne LL, Tatpati AD, Lynch HT, Watson P. Histology of prophylactically removed ovaries from BRCA1 and BRCA2 mutation carriers compared with noncarriers in hereditary breast ovarian cancer syndrome kindreds.
Gynecol Oncol
2000
;
78
:
278
–87.
8
Piek JM, van Diest PJ, Zweemer RP, et al. Dysplastic changes in prophylactically removed Fallopian tubes of women predisposed to developing ovarian cancer.
J Pathol
2001
;
195
:
451
–6.
9
Medeiros F, Muto MG, Lee Y, et al. The tubal fimbria is a preferred site for early adenocarcinoma in women with familial ovarian cancer syndrome.
Am J Surg Pathol
2006
;
30
:
230
–6.
10
Finch A, Shaw P, Rosen B, Murphy J, Narod SA, Colgan TJ. Clinical and pathologic findings of prophylactic salpingo-oophorectomies in 159 BRCA1 and BRCA2 carriers.
Gynecol Oncol
2006
;
100
:
58
–64.
11
Crum CP, Drapkin R, Kindelberger D, Medeiros F, Miron A, Lee Y. Lessons from BRCA: the tubal fimbria emerges as an origin for pelvic serous cancer.
Clin Med Res
2007
;
5
:
35
–44.
12
Piek JM, Kenemans P, Verheijen RH. Intraperitoneal serous adenocarcinoma: a critical appraisal of three hypotheses on its cause.
Am J Obstet Gynecol
2004
;
191
:
718
–32.
13
Lee Y, Miron A, Drapkin R, et al. A candidate precursor to serous carcinoma that originates in the distal fallopian tube.
J Pathol
2007
;
211
:
26
–35.
14
Kindelberger DW, Lee Y, Miron A, et al. Intraepithelial carcinoma of the fimbria and pelvic serous carcinoma: evidence for a causal relationship.
Am J Surg Pathol
2007
;
31
:
161
–9.
15
Crum CP, Drapkin R, Miron A, et al. The distal fallopian tube: a new model for pelvic serous carcinogenesis.
Curr Opin Obstet Gynecol
2007
;
19
:
3
–9.
16
Marquez RT, Baggerly KA, Patterson AP, et al. Patterns of gene expression in different histotypes of epithelial ovarian cancer correlate with those in normal fallopian tube, endometrium, and colon.
Clin Cancer Res
2005
;
11
:
6116
–26.
17
Riska A, Sund R, Pukkala E, Gissler M, Leminen A. Parity, tubal sterilization, hysterectomy and risk of primary fallopian tube carcinoma in Finland, 1975–2004.
Int J Cancer
2007
;
120
:
1351
–4.
18
Pectasides D. Fallopian tube carcinoma: a review.
The Oncologist
2006
;
11
:
902
–12.
19
Pere H, Tapper J, Seppala M, Knuutila S, Butzow R. Genomic alterations in fallopian tube carcinoma: comparison to serous uterine and ovarian carcinomas reveals similarity suggesting likeness in molecular pathogenesis.
Cancer Res
1998
;
58
:
4274
–6.
20
Nowee M, Snijders A, Rockx D, et al. DNA profiling of primary serous ovarian and Fallopian tube carcinomas with array comparative genomic hybridization and multiplex ligation-dependent probe amplification.
J Pathol
2007
;
213
:
46
–55.
21
Dafforn A, Chen P, Deng G, et al. Linear mRNA amplification from as little as 5 ng total RNA for global gene expression analysis.
BioTechniques
2004
;
37
:
3
–7.
22
Sultan M, Wigle DA, Cumbaa CA, et al. Binary tree-structured vector quantization approach to clustering and visualizing microarray data.
Bioinformatics
2002
;
18
Suppl 1:
S111
–9.
23
Brown KR, Jurisica I. Unequal evolutionary conservation of human protein interactions in interologous networks.
Genome Biol
2007
;
8
:
R95
.
24
Camp RL, Charette LA, Rimm DL. Validation of tissue microarray technology in breast carcinoma.
Lab Invest
2000
;
80
:
1943
–9.
25
Narod SA, Foulkes WD. BRCA1 and BRCA2: 1994 and Beyond.
Nat Rev Ca
2004
;
4
:
665
–76.
26
Boulton SJ. Cellular functions of the BRCA tumour-suppressor proteins.
Biochem Soc Trans
2006
;
34
:
633
–45.
27
Yoshida K, Miki Y. Role of BRCA1 and BRCA2 as regulators of DNA repair, transcription, and cell cycle in response to DNA damage.
Cancer Sci
2004
;
95
:
866
–71.
28
Verhage HG, Bareither ML, Jaffe RC, Akbar M. Cyclic changes in ciliation, secretion and cell height of the oviductal epithelium in women.
Am J Anat
1979
;
156
:
505
–21.
29
Verhage HG, Fazleabas AT, Mavrogianis PA, et al. Characteristics of an oviductal glycoprotein and its potential role in fertility control.
J Reprod Fertil Suppl
1997
;
51
:
217
–26.
30
Rosen EM, Fan S, Isaacs C. BRCA1 in hormonal carcinogenesis: basic and clinical research.
Endocr Relat Cancer
2005
;
12
:
533
–48.
31
Ma Y, Katiyar P, Jones LP, et al. The breast cancer susceptibility gene BRCA1 regulates progesterone receptor signaling in mammary epithelial cells.
Mol Endocrinol
2006
;
20
:
14
–34.
32
Zheng H, Kavanagh JJ, Hu W, Liao Q, Fu S. Hormonal therapy in ovarian cancer.
Int J Gynecol Cancer
2007
;
17
:
325
–38.
33
Risch HA. Hormonal etiology of epithelial ovarian cancer, with a hypothesis concerning the role of androgens and progesterone.
J Natl Cancer Inst
1998
;
90
:
1774
–86.
34
Ho SM. Estrogen, progesterone and epithelial ovarian cancer.
Reprod Biol Endocrinol
2003
;
1
:
73
.
35
Prat J. Hereditary ovarian cancer.
Hum Pathol
2005
;
36
:
861
–70.
36
Schildkraut JM, Calingaert B, Marchbanks PA, Moorman PG, Rodriguez GC. Impact of progestin and estrogen potency in oral contraceptives on ovarian cancer risk.
J Natl Cancer Inst
2002
;
94
:
32
–8.
37
McDonnel AC, Van Kirk EA, Isaak DD, Murdoch WJ. Inhibitory effects of progesterone on plasma membrane fluidity and tumorigenic potential of ovarian epithelial cancer cells.
Exp Biol Med (Maywood)
2003
;
228
:
308
–14.
38
Syed V, Ulinski G, Mok SC, Yiu GK, Ho SM. Expression of gonadotropin receptor and growth responses to key reproductive hormones in normal and malignant human ovarian surface epithelial cells.
Cancer Res
2001
;
61
:
6768
–76.
39
Poole AJ, Li Y, Kim Y, Lin SC, Lee WH, Lee EY. Prevention of Brca1-mediated mammary tumorigenesis in mice by a progesterone antagonist.
Science
2006
;
314
:
1467
–70.
40
Shah A, Nandedkar TD, Raghavan VP, Parulekar SV, Natraj U. Characterization and localization of estrogen and progesterone receptors of human fallopian tube.
Indian J Exp Biol
1999
;
37
:
893
–99.
41
Khan MS, Heatley MK. The expression of the proliferation marker Ki-67 and oestrogen and progesterone receptors in the proximal and distal fallopian tube.
J Obstet Gynaecol
1999
;
19
:
519
–21.
42
Rzepka-Gorska I, Tarnowski B, Chudecka-Glaz A, Gorski B, Zielinska D, Toloczko-Grabarek A. Premature menopause in patients with BRCA1 gene mutation.
Breast Cancer Res Treat
2006
;
100
:
59
–63.
43
Reedy MB, Hang T, Gallion H, Arnold S, Smith SA. Antisense inhibition of BRCA1 expression and molecular analysis of hereditary tumors indicate that functional inactivation of the p53 DNA damage response pathway is required for BRCA-associated tumorigenesis.
Gynecol Oncol
2001
;
81
:
441
–6.
44
Hocevar BA, Smine A, Xu XX, Howe PH. The adaptor molecule disabled-2 links the transforming growth factor β receptors to the Smad pathway.
EMBO J
2001
;
20
:
2789
–801.
45
Yamada SD, Baldwin RL, Karlan BY. Ovarian carcinoma cell cultures are resistant to TGF-β1-mediated growth inhibition despite expression of functional receptors.
Gynecol Oncol
1999
;
75
:
72
–7.
46
He J, Tegen SB, Krawitz AR, Martin GS, Luo K. The transforming activity of Ski and SnoN is dependent on their ability to repress the activity of Smad proteins.
J Biol Chem
2003
;
278
:
30540
–7.
47
Karlsson G, Liu YJ, Larsson J, et al. Gene expression profiling demonstrates that TGF-β1 signals exclusively through receptor complexes involving Alk5 and identifies targets of TGF-β signaling.
Physiol Genomics
2005
;
21
:
396
–403.
48
Levy L, Hill CS. Smad4 dependency defines two classes of transforming growth factor β (TGF-β) target genes and distinguishes TGF-β-induced epithelial-mesenchymal transition from its antiproliferative and migratory responses.
Mol Cell Biol
2005
;
25
:
8108
–25.
49
Ranganathan P, Agrawal A, Bhushan R, et al. Expression profiling of genes regulated by TGF-β: differential regulation in normal and tumour cells.
BMC Genomics
2007
;
8
:
98
–116.
50
Massague J, Gomis RR. The logic of TGFβ signaling.
FEBS Lett
2006
;
580
:
2811
–20.
51
Yarden RI, Pardo-Reoyo S, Sgagias M, Cowan KH, Brody LC. BRCA1 regulates the G2/M checkpoint by activating Chk1 kinase upon DNA damage.
Nat Genet
2002
;
30
:
285
–9.
52
He J, Xu J, Xu XX, Hall RA. Cell cycle-dependent phosphorylation of disabled-2 by cdc2.
Oncogene
2003
;
22
:
4524
–30.
53
Yang DH, Fazili Z, Smith ER, et al. Disabled-2 heterozygous mice are predisposed to endometrial and ovarian tumorigenesis and exhibit sex-biased embryonic lethality in a p53-null background.
Am J Pathol
2006
;
169
:
258
–67.
54
Santin AD, Zhan F, Bellone S, et al. Gene expression profiles in primary ovarian serous papillary tumors and normal ovarian epithelium: identification of candidate molecular markers for ovarian cancer diagnosis and therapy.
Int J Cancer
2004
;
112
:
14
–25.
55
Mok SC, Chan WY, Wong KK, et al. DOC-2, a candidate tumor suppressor gene in human epithelial ovarian cancer.
Oncogene
1998
;
16
:
2381
–7.
56
Zhang F, Lundin M, Ristimaki A, et al. Ski-related novel protein N (SnoN), a negative controller of transforming growth factor-β signaling, is a prognostic marker in estrogen receptor-positive breast carcinomas.
Cancer Res
2003
;
63
:
5005
–10.
57
Krakowski AR, Laboureau J, Mauviel A, Bissell MJ, Luo K. Cytoplasmic SnoN in normal tissues and nonmalignant cells antagonizes TGF-β signaling by sequestration of the Smad proteins.
Proc Natl Acad Sci U S A
2005
;
102
:
12437
–42.
58
Zhu Q, Krakowski AR, Dunham EE, et al. Dual role of SnoN in mammalian tumorigenesis.
Mol Cell Biol
2007
;
27
:
324
–39.

Supplementary data