Skip to main content
Erschienen in: Journal of Ovarian Research 1/2016

Open Access 01.12.2016 | Research

Molecular manipulation of keratin 8/18 intermediate filaments: modulators of FAS-mediated death signaling in human ovarian granulosa tumor cells

Erschienen in: Journal of Ovarian Research | Ausgabe 1/2016

Abstract

Background

Granulosa cell tumors (GCT) are a rare ovarian neoplasm but prognosis is poor following recurrence. Keratin intermediate filaments expressed in these tumors are a diagnostic marker, yet paradoxically, may also constitute a target for therapeutic intervention. In the current study, we evaluated keratin 8/18 (K8/18) filament expression as a mechanism of resistance to apoptosis in GCT, specifically focusing on regulation of the cell surface death receptor, Fas (FAS).

Methods

The GCT cell line, KGN, was transiently transfected with siRNA to KRT8 and KRT18 to reduce K8/18 filament expression. Expression of K8/18, FAS, and apoptotic proteins (PARP, cleaved PARP) were evaluated by fluorescence microscopy, flow cytometric analysis, and immunoblotting, respectively. The incidence of FAS-mediated apoptosis in KGN cells was measured by caspase 3/7 activity. All experiments were performed independently three to six times, using a fresh aliquot of KGN cells for each experiment. Quantitative data were analyzed by one- or two-way analysis of variance (ANOVA), followed by a Tukey’s post-test for multiple comparisons; differences among means were considered statistically significant at P < 0.05.

Results

Control cultures of KGN cells exhibited abundant K8/18 filament expression (~90 % of cells), and minimal expression of FAS (<25 % of cells). These cells were resistant to FAS-activating antibody (FasAb)-induced apoptosis, as determined by detection of cleaved PARP and measurement of caspase 3/7 activity. Conversely, siRNA-mediated knock-down of K8/18 filament expression enhanced FAS expression (> 70 % of cells) and facilitated FasAb-induced apoptosis, evident by increased caspase 3/7 activity (P < 0.05). Additional experiments revealed that inhibition of protein synthesis, but not MEK1/2 or PI3K signaling, also prompted FasAb-induced apoptosis.

Conclusions

The results demonstrated that K8/18 filaments provide resistance to apoptosis in GCT by impairing FAS expression. The abundance of keratin filaments in these cells and their role in apoptotic resistance provides a greater mechanistic understanding of ovarian tumorgenicity, specifically GCT, as well as a clinically-relevant target for potential therapeutic intervention.
Hinweise

Competing interests

The authors declare they have no competing interests.

Author’s contributions

SKT, NMS, and XH performed the experiments, conducted the statistical analyses, and assisted in the preparation of the manuscript. SKT, JSD and DHT conceived the study, participated in its design and coordination, and wrote the manuscript. All authors read and approved the final manuscript.
Abkürzungen
CASP8
caspase 8
CFLAR
CASP8 and FADD-like apoptosis regulator
CHX
cycloheximide
DAPI
4′,6-diamidino-2-phenylindole
DISC
death-inducing signaling complex
FAS
Fas cell surface death receptor (also known as TNFRSF6)
FasAb
FAS-activating antibody
FASL
FAS ligand
FITC
Fluorescein isothiocyanate
FSH
follicle-stimulating hormone
GCT
granulosa cell tumor
K
keratin (protein)
KRT
keratin (DNA/RNA)
MEK1/2
Mitogen-activated protein kinase kinase 1 and 2
NGS
normal goat serum
OSE
ovarian surface epithelium
PARP
Poly (ADP-ribose) polymerase
PI3K
Phosphatidylinositol-4,5-bisphosphate 3-kinase
siRNA
short interfering RNA
TLR
Toll-like receptor
TNFRSF1
Tumor necrosis factor receptor 1
TRAIL
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand

Background

Granulosa cell tumors (GCT) represent approximately 5 % of ovarian malignancies, yet constitute the most prevalent sex cord-stromal ovarian neoplasms [1, 2]. Prognosis is favorable when detected early, but recurrent GCT may be life-threatening. The rarity of these tumors and their low potential for malignancy (i.e., slow growth over a prolonged period) has reduced the urgency to study and develop standard treatment regimens for these neoplasms compared to other ovarian cancers. However, GCT have a high rate of reappearance following resection [24], and such recurrence often leads to late detection and poor prognosis [5, 6]. Distinct from other ovarian cancers, GCT are endocrine tumors [7]. High estrogen secretion by GCT can lead to a variety of malignancies and pathological states, including oligomenorrhea, hirsutism, breast cancer, endometrial hyperplasia and adenocarcinoma [5, 810]. Additionally, GCT have many morphological and biochemical features akin to granulosa cells of mature, preovulatory follicles (e.g., FSH-responsive and steroid hormone production) [1114].
Tumorigenicity of GCT is linked to a somatic missense mutation in the Forkhead Box L2 (FOXL2) gene (c.402C→G; p.C134W). The mutation occurs in nearly all adult-type GCT, yet is absent in juvenile GCT [14], indicating the FOXL2 mutation is etiologically significant in the development of adult GCT. The function of the mutated FOXL2 is not fully understood, however it is postulated to be a tumor suppressor. Overexpression of FOXL2 induces expression of cell death receptors of the Tumor Necrosis Factor Receptor Superfamily, particularly the cell surface death receptor, Fas (FAS, also known as TNFRSF6), which facilitates apoptosis of ovarian granulosa cells [15, 16]. In contrast, granulosa cells expressing the FOXL2 C134W mutant lack these death receptors and are resistant to apoptosis [16]. In spite of recent attempts to develop therapeutic approaches using genetic manipulation in mouse models [14, 17, 18] and transcriptomic analysis to identify candidate driver genes [19, 20], relatively little is known about selectively treating GCT.
Current treatment of GCT entails surgical resection of the ovary and/or platinum-based chemotherapy - originally developed to specifically eliminate ovarian surface epithelial (OSE) tumors [21]. However, ovarian carcinomas, including GCT and OSE tumors, share the common trait of expressing keratin intermediate filaments. Keratin type I cytoskeletal 18 protein (K18, also known as KRT18), in particular, is expressed in malignant cells and is a diagnostic marker to delineate morphologic heterogeneity and neoplastic changes [22, 23]. Others argue that the prevalence of intermediate filaments (i.e., keratins, vimentin, and desmin) in GCT, as well as other ovarian carcinomas, limits their value in differential diagnosis among these groups [24].
The well-understood function of keratin intermediate filaments is to provide stability and morphological integrity to cells, yet in recent years a more dynamic role has emerged. Structurally, keratin filaments are heterotetramers of two type I and two type II keratins. Keratin 8/18 (K8/K18) intermediate filaments, for example, are composed of keratin type II cytoskeletal 8 protein and type I cytoskeletal 18 protein. Originally named for their mechanical stability, K8/18 filaments are “stress filaments” that provide resistance against sheer stress; but they also influence intracellular signaling mechanisms, counteract physiological stressors, enhance wound healing, and prevent apoptosis [25, 26]. Notably, K8/18-deficient epithelial cells are sensitive to physiological stressors, including cytokine-induced apoptosis [2730]. In this context, K8/18 filaments within cells associate with the tumor necrosis factor receptor 1-associated death domain (TRADD) protein [31]. TRADD is a critical intracellular intermediate to TNFR1-induced death signaling, which in turn interacts with the Fas-Associated Death Domain (FADD) protein [32]. By sequestering TRADD, K8/18 filaments potentially attenuate and influence interactions between FAS, FADD and other downstream apoptotic molecues, thus providing resistance to cytokine-mediated apoptosis. In carcinoma-derived cell lines (HeLa, HepG2, KLE), for example, disruption of K8/18 filament expression sensitizes the cells to cisplatin-induced apoptosis by increasing FAS expression, but also by decreasing the expression of the anti-apoptotic protein, cellular FLICE inhibitory protein (c-FLIP, also known as CFLAR) [33]. Based upon these observations, keratin filaments constitute a plausible target for immune-based approaches to cancer treatment, and might provide an alternative to chemotherapy of GCT, in particular.
The granulosa-like KGN cell line, established from an adult GCT, has provided insight about the molecular and cellular features of GCT, along with potential vision for treatment [34]. For instance, a recent investigation revealed that, in contrast to OSE-derived cancers, inhibition of NF-κB does not sensitize KGN cells to TRAIL- or cisplatin-induced apoptosis [35]. Interestingly, human granulosa cells and KGN cells share a similar resistance to other forms of immune-mediated death, including FAS-induced apoptosis [34, 36, 37]. Acknowledging that K8/18 filament loss in other epithelial cell types leads to increased FAS expression and enhanced vulnerability to FAS-mediated apoptosis [28, 33], we hypothesized in the current study, that K8/18 filaments in KGN cells provide a similar mechanism of influence. Our objectives were to first determine if KGN cells express K8/18 filaments as seen in granulosa cells, GCT, and OSE tumors [22, 23, 30] and then to identify the cellular mechanism(s) by which K8/18 filaments potentially influence resistance to FAS-mediated apoptosis. We observed that, indeed, KGN cells express an abundance of K8/18 filaments and genetic knockdown of K8/18 results in enhanced expression of FAS and increased vulnerability to FAS-mediated apoptosis.

Methods

Cells and culture conditions

The human granulosa cell tumor line, KGN, was generously provided by Dr. Fukuzawa (RIKEN Cell Bank, Koyadai, Japan). KGN cells were maintained in DMEM/F12 supplemented with 10 % FBS at 37 °C with 5 % CO2, 95 % air and 95 % humidity. Cells were seeded in multi-well plates, T25-flasks (Corning, Corning, NY) or microchamber slides (Nunc, Rochester, NY) at a relative seeding density of 5 × 104 cells/mL and grown to 70 % confluency prior to treatment.

Immunofluorescent visualization of K8/K18 intermediate filaments

The K18 protein is known to dimerize with K8 protein to form K8/K18 filaments in ovarian steroidogenic cells [30, 38]. KGN cells grown on coverslips were washed with PBS, then fixed with ice-cold 4 % paraformaldehyde (PFA) in PBS (pH 7.4) for 10 min. The fixed cells were permeabilized for 10 min in 0.4 % Triton-X-100 in PBS. Cells were washed and blocked with blocking buffer (0.2 % Triton-X-100 in PBS with 10 % normal donkey serum) for 30 min at room temperature. Cells were labeled with mouse anti-human K18-FITC-conjugated antibody (CY90; Sigma-Aldrich, St. Louis, MO) diluted 1:100 in blocking buffer for 3.5 h at room temperature. Cells were washed again and probed with Rhodamine Phalloidin (1:1500, Life Technologies, Grand Island, NY) and 4′,6-diamidino-2-phenylindole (DAPI; 30 nM) in blocking buffer for 30 min at room temperature. For the negative control, KGN cells were also exposed to a mouse anti- human IgG-FITC-conjugated antibody (Sigma-Aldrich, St. Louis, MO) diluted 1: 100 in blocking buffer in place of the primary. The coverslips were washed and then mounted with Fluoromount-G® (Southern Biotechnology Associates, Inc, (Birmingham, AL). Images were captured using a Zeiss 710 Meta Confocal Laser Scanning Microscope and analyzed using the Zeiss Zen 2010 software (Carl Zeiss Microscopy, LLC, Thornwood, NY, USA).

Flow cytometric quantification of FAS surface and K18 expression

Cultured cells were trypsinized using Cellgro 0.25 % Trypsin-EDTA (Corning, Corning, NY) and fixed with 2 % PFA in microtubule-stabilizing buffer. For FAS staining, the cells were stored in PFA; for K18 and total FAS staining, the fixed cells were permeabilized in 70 % ethanol. For antibody labeling, both types of cell preparations were washed with PBS + 0.1 % BSA, and then either labeled overnight with mouse anti-human FAS (CH11; EMD Millipore, Darmstadt, Germany) antibody diluted to 20 μg/mL in 10 % normal goat serum (NGS) + PBS + 1 % BSA, or for 1 h with mouse anti-human K18-FITC-conjugated antibody (Sigma-Aldrich, St. Louis, MO) diluted 1:100 in PBS + 1 % BSA. Separate fixed cell suspensions were labeled with mouse anti-human IgG-FITC-conjugated antibody (Sigma-Aldrich, St. Louis, MO) to serve as the negative control. For FAS staining, the cells were subsequently washed with PBS + 0.1 % BSA, and incubated with Alexa Fluor® 488 (Life Technologies, Grand Island, NY) diluted 1:200 in 10 % NGS + PBS + 1 % BSA. All samples were washed with PBS + 0.1 % BSA and analyzed using a 4 color, dual laser FACS Calibur Flow Cytometer (BD Biosciences, San Jose, CA), quantifying 10,000 cells for each sample. Data were collected using CellQuest software (BD Biosciences; San Jose, CA) and then analyzed using WinMDI (Joe Trotter; Purdue University, West Lafayette, IN) to quantify FAS and keratin filament expression.

Induction of FAS-mediated apoptosis

KGN cells were pretreated with cycloheximide (CHX; 0.25 μg/mL; Sigma-Aldrich, St. Louis, MO), the MEK1/2 inhibitor PD98059 (30 μM; Cell Signaling Technologies, Danvers, MA) or the PI3K inhibitor Wortmannin (100 nM; EMD Millipore, Darmstadt, Germany) for 2 h in serum-free culture medium. After pretreatment, the cells were exposed to FAS-activating antibody (FasAb; 1 μg/mL; clone CH11; EMD Millipore, Darmstadt, Germany), or staurosporine (1 μM; MP Biomedical, Santa Ana, CA) as a positive control, to induce apoptosis. The cells were exposed to the above treatments for 8 and 24 h, at which time caspase 3/7 activity was measured by the Caspase-Glo® 3/7 Assay and cell viability was determined using the CellTiter 96® AQueous One Solution Cell Proliferation Assay (MTS), respectively. Assays were conducted according to the manufacturer’s instructions (Promega, Madison, WI).

Immunoblot analysis for FAS, cFLIP, cleaved PARP and β-actin

Nearly confluent cells from the above-described experiments were washed with PBS and harvested in lysis buffer (10 mM Tris–HCl; 1 mM EDTA; 1 mM EGTA; 100 mM NaCl; 1 % Triton X-100; 0.5 % Nonidet P-40, pH 7.4) containing a cocktail of kinase, protease and phosphatase inhibitors (Sigma-Alrich, St. Louis, MO). Cells were scraped, collected, sonicated and then resuspended in 2X SDS loading buffer (100 mM Tris-Cl, pH 6.8 + 4 % SDS, 0.2 % bromophenol blue, 20 % glycerol, 200 mM DTT) before denaturing at 95 °C for 5 min. Total cellular proteins were separated by SDS-PAGE and transferred to polyvinylidene difluoride (PVDF) membranes (EMD Millipore,Darmstadt, Germany). Immunoblotting was performed using antibodies to detect human cFLIP (also known as CFLAR) (rabbit anti-human CFLAR; Sigma Aldrich, St. Louis, MO) and human FAS (clone C-20; Santa Cruz Biotechnology, Santa Cruz, CA). The membranes were stripped and reprobed for cleaved human poly ADP ribose polymerase (PARP) (# 9542, Cell Signaling Technology, Danvers, MA), to verify the apoptotic effect, and β-actin (clone AC-15; Sigma Aldrich, St. Louis, MO) to ensure equivalent protein loading.

Short interfering RNA (siRNA) knockdown of KRT8/18 gene transcription

KGN cells were transiently transfected with 10 pmol siRNA constructs to KRT8 and KRT18 (Smartpool Accell KRT8 and KRT18 siRNA; Dharmacon RNAi, GE Healthcare, Lafayette, CO). Transfection was achieved using Lipofectamine™ RNAiMAX in OptiMEM® Reduced Serum Media for final 100 nM RNAi duplexes according to the manufacturer’s instructions (Life Technoloies, Grand Island, NY). Cells were grown to 70 % confluency then switched to antibiotic-free DMEM/F12 + 10 % FBS before siRNA- Lipofectamine™ duplexes were introduced. The cells were also exposed to Lipofectamine™ and a non-targeting siRNA (siCTL; Silencer® Select Negative Control #1; Ambion Inc., Foster City, CA) as negative controls. Knock-down of KRT8/18 expression was evaluated by immunofluorescence as described above.
Knock-down of KRT8/18 was also evaulated using an in-cell western assay according to the manufacturer’s instructions (LI-COR®, Lincoln, NE). Following a 72 h exposure to siRNA, the cells were washed with PBS, then fixed and permeabilized in 100 % MeOH. Detection of K18 and β-Actin (internal control) expression was achieved using an antibody cocktail of mouse anti-human K18 (CY90; Sigma-Aldrich, St. Louis, MO) and rabbit anti-human β-Actin (13E5, Cell Signaling Technolgy, Danvers, MA) followed by a secondary antibody cocktail (goat anti-mouse IgG H + L DyLight 800 and goat anti-rabbit IgG H + L DyLight 680, Cell Signaling Technology, Danvers MA). The cells were imaged using a LI-COR® Odyssey® Classic Infrared Imaging scanner. Staining intensity for K18 was normalized to the staining intensity for β-actin using the provided software.

Statistical analysis

All experiments were independently replicated three to six times, using a fresh aliquot of KGN cells (passage 23-27) to initiate each experiment. Data were analyzed by one- or two-way analysis of variance (ANOVA), followed by a Tukey’s post-test for multiple comparisons; differences among means were considered statistically significant at P < 0.05.

Results

Keratin and β-actin expression in KGN cells

Immunofluorescent staining of cultured KGN cells revealed an abundance of K8/18 filament expression throughout the cells (Fig. 1, green fluorescence). Both cytoplasmic and perinuclear expression was evident, generally in parallel with β-actin filament expression (Fig. 1, red fluorescence). Thus, both K8/18 and β-actin filament expression are prominent cytoskeletal components of cultured KGN cells.

Abundant K18 expression but minimal FAS expression on KGN Cells

Flow cytometric analysis demonstrated ~ 91 % of KGN cultured cells expressed K18 (Fig. 2a and c); consistent with the abundance of keratin filament staining observed by immunofluorescence (representative image shown in Fig. 1). In contrast, only 24 % of the cells expressed FAS (Fig. 2b and c), indicative of an overall inverse relationship between K8/18 filament abundance and FAS expression. Mean fluorescent intensity measures (an indication of fluorescence per cell) were similarly high for keratin filament expression, but low for FAS expression (Fig. 2d).

Inhibition of protein synthesis sensitizes KGN cells to FasAb-induced apoptosis

Exposure of KGN cells to FasAb for 8 h failed to induce apoptosis, as measured by caspase 3/7 activity (Fig. 3). Conversely, pretreatment with cycloheximide (CHX, 0.25 μg/ml), a protein synthesis inhibitor, augmented FasAb-induced apoptosis (P < 0.05; Fig. 3). The effect was similar to that of Staurosporine (1 μM), a chemotherapeutic agent used as a positive control (P < 0.05, Fig. 3). Exposure of KGN cells to CHX alone had no effect (P > 0.05). Similarly, disruption of MEK1/2 (via PD98059; 30 μM) and PI3K (via Wortmannin; 100 nM) pro-survival pathways in KGN cells, failed to augment FasAb-induced apoptosis (data not shown).

FAS and cFLIP expression following FasAb-induced apoptosis in KGN cells

Immunoblotting revealed no overt changes in FAS expression following exposure to CHX or FasAb in cultured KGN cells (P > 0.05, Fig. 4a and b). Similarly, there was no effect of CHX and/or FasAb on the expression of cFLIP (P > 0.05, Fig. 4a and c). In contrast, effects of these treatments were evident by the detection of PARP and cleaved PARP (Fig. 4a and d), but only the combined treatment led to a doubling (P < 0.05) of cleaved PARP (Fig. 4d). The results confirmed the combined apoptotic effects of CHX and FasAb as shown in Fig. 3.

siRNA knockdown of KRT8 and KRT18 enhances FasAb-induced apoptosis of KGN cells

Knockdown of KRT8 and KRT18 in KGN cells (siRNA) reduced keratin expression by 63 % as determined by in-cell western (Fig. 5a and b) and 72 % by confocal imaging (Fig. 5c). Subsequent experiments evaluated the effect of KRT8 and KRT18 knockdown on FasAb-induced apoptosis in KGN cells. Treatment with Lipofectamine™, non-targeting siRNA (siCTL) or KRT8/18 siRNA alone had no effect on caspase 3/7 activity (P > 0.05, Fig. 6); however, subsequent exposure to FasAb for 8 h resulted in a higher incidence of apoptosis in KRT8/18 siRNA-treated cells compared to controls (P < 0.05; Fig. 6). Of interest, treatment with Lipofectamine™ and siCTL moderately augmented the sensitivity of KGN cells to the apoptosis-inducing effects of FasAb (Fig. 6).

Down-regulation of keratin filaments (siRNA) enhances expression of FAS on KGN cells

Additional experiments revealed that siRNA-mediated reduction of K8/18 expression enhanced expression of FAS on the surface of KGN cells by 21 % as determined by flow cytometry (P < 0.05; Fig. 7). By comparison, no difference in total FAS expression (P > 0.05) was observed among the treatment groups (Fig. 7). Of note, Lipofectamine™ alone elevated the percentage of KGN cells that expressed FAS on the cell surface (58 %, Fig. 7) compared to that observed previously in non-treated controls (24 %, Fig. 2). However, the effect did not overall enhance the sensitivity of the cells to FasAb-induced apoptosis (P > 0.05 Fig. 6).

Discussion

The current study is the first to demonstrate the expression of K8/18 filaments in KGN cells, a GCT-derived cell line, which has clinical relevance to GCT in particular, but also to general ovarian malignancy. Acknowledging that these intermediate filaments help to counteract physiological stressors [28, 39], we show that disrupting K8/18 filament expression (via siRNA) in KGN cells reverses resistance to FAS-mediated apoptosis by enhancing FAS expression on the cell surface
Keratin filaments are typically expressed by cells of epithelial origin [40]. Although the composition of keratin filaments can be quite diverse and is predicated by cell- and tissue-origin, previous reports document the assembly of K18-containing filaments within ovarian-derived tissues and tumors [26, 30, 38, 41]. Keratin filaments constitute one type of intermediate filament detectable within the ovary, particularly in granulosa cells of the follicle during stages of growth and atresia [30, 42], in luteal cells of the corpus luteum throughout the luteal phase [30, 38, 43], and in oocytes of fetal and adult ovaries [44, 45]. Other types of intermediate filaments expressed within cells of the ovary include vimentin and desmin [42, 46], which are of mesenchymal and myogenic origins, respectively [46]. Whether or not intermediate filaments other than K8/18 filaments impact FAS expression or FAS-mediated apoptosis is unknown.
FAS-mediated apoptosis is characterized by the localized secretion of Fas ligand, which then binds to FAS on the target cell, inducing oligomerization of the ligand-receptor complex. Oligomerization and internalization of the ligand-receptor complex triggers association with the cytoplasmic Fas-Associated Death Domain (FADD) protein [47]. Interestingly, FADD is the common point of convergence for both TNFR1-induced (via TRADD) and Fas ligand-induced pathways of apoptosis in cells [32]. In the current study, however, we focused only on the FAS-mediated pathway of cell death. Subsequent to activation, FADD facilitates the binding of pro-caspase 8 and the formation of the death inducing signaling complex (DISC). Assembly of the DISC results in cleavage of pro-caspase 8 to activate caspase 8, and initiates downstream events including the activation of caspase 3/7 enzymes that ultimately trigger apoptosis of the cell [48].
In the context of GC and GCT, the expression of FAS fundamentally determines the relative sensitivity of the cells to FAS-mediated apoptosis. Overexpression of wild-type, but not mutant FOXL2, in GC for example, upregulates FAS expression and augments Fas ligand-induced apoptosis [16]. Others report that granulosa and KGN cells normally resistant to Fas ligand-induced apoptosis become sensitized when pre-exposed to cytokines that up-regulate FAS expression, such as interferon gamma and tumor necrosis factor alpha [34, 37, 49]. In the present study we show for the first time that siRNA-mediated knockdown of K8/18 filaments in these cells also increases FAS expression and enhances FasAb-induced apoptosis. Consistent with the concept that FAS expression ultimately determines the sensitivity to FAS-mediated apoptosis, we also report that treatment of KGN cells with Lipofectamine™ increases trafficking of FAS to the cell surface and enhances responsiveness to FasAb, albeit to a lesser extent than the depletion of K8/18 filaments.
Inhibition of protein synthesis (via CHX) similarly enhances cell sensitivity to FasAb-induced apoptosis [36], but does so ostensibly by impairing the synthesis of cytoplasmic and labile, anti-apoptotic proteins rather than influencing FAS expresssion. In the current study, we observed increased apoptosis of KGN cells (i.e., increased caspase 3/7 activity and cleaved PARP expression) only after pretreating the cells with CHX for 2 h and then exposing them to FasAb (Figs.  3 and 4 ). Conversely, treatment with CHX or FasAb alone failed to induce apoptosis, and there was no effect of either treatment, alone or in combination, on the total expression of FAS or cFLIP (Fig.  4 ), a well-documented anti-apoptotic protein [50]. It is unclear why in the current study transient inhibition of protein synthesis failed to reduce cFLIP expression, yet augmented FasAb-induced apoptosis. Others have suggested that cFLIP plays an important role in preventing apoptosis in KGN cells [36, 51] and that CHX inhibits cFLIP expression [36, 52]. One explanation for the discrepancy in results between these previous reports and the current study might be the concentration of CHX used to inhibit protein synthesis (5-25 μg/mL in previous reports versus 0.25 μg/mL in this study, respectively). Isoforms of cFLIP also exist [5355], which further complicates the interpretation of direct comparisons among studies. For these reasons, we suggest that additional investigation is needed to clarify the role of cFLIP as well as other potential anti-apoptotic molecules in KGN cells.
The activation of MAPK/ERK1/2 and PI3K/AKT signaling pathways have been implicated in the proliferation of KGN cells [56]. In previous work, we determined that inhibitors of these pro-survival signaling pathways prevent TGFα-induced phosphorylation of ERK1/2 and AKT in KGN cells [57], providing a potential role for TGFα in GCT initiation, progression and metastasis. However, inhibition of ERK1/2 phosphorylation has little to no effect on cell cycle progression or proliferation of KGN cells. Conversely, blocking AKT activation, or its downstream target mTOR, impairs cell cycle progression and prevent cell proliferation in cell culture [57] and in vivo models of GCT [58]. Thus, the AKT pathway appears more influential on the tumorigenic potential of KGN cells than the ERK1/2 pathway, and this is consistent with the identification of AKT1 as a driver gene of GCTs [19]. In the present study, we observed that inhibiting either the ERK1/2 or AKT pathway (via PD98059 and Wortmannin, respectively) failed to augment the apoptotic effects of FasAb, as suggested by other models [5961]. In the context of the present study, keratin filaments are known to regulate cell cycle progression [62, 63]. Therefore, we speculate that these filaments have the potential to modulate the effects of cell signaling inhibitors on KGN cell proliferation and apoptotic sensitivity.
The concept that keratin filaments influence the trafficking of cytokine receptors, as demonstrated in the current study, is a relatively new and unique attribute ascribed to intermediate filaments. Inhibition of keratin filament expression deems epithelial cells and carcinoma cell lines more sensitive to FAS-mediated apoptosis via a receptor mechanism [64, 65]. Additionally, expression of keratin filaments within a lymph node metastatic carcinoma cell line impairs human leukocyte antigen (HLA) class I receptor expression, enabling the cells to avoid destruction by CD8+ cytotoxic lymphocytes [66]. Our observations in which siRNA-mediated knockdown of K8/18 filaments in KGN cells increased FAS expression and enhanced sensitivity to FasAb-induced apoptosis is consistent with evidence that shows knockdown of KRT18 increases FAS expression in both non-tumorigenic and tumorigenic cells [28, 33]. The abundance of keratin filaments in tumorigenic cells and their influence on metastatic potential has generated considerable interest among oncologists in recent years [6769]. The current thinking is that keratin filaments facilitate migration of cancer cells in a manner separate from epithelial-mesenchymal transitions. Yet, surprisingly, this role of keratin filaments in cancer progression also renders some cell-types more sensitive to cisplatin therapy [33]. Of relevance to GCT, Woods and co-workers determined that KGN cells are resistant to cisplatin and tumor necrosis factor-related apoptosis inducing ligand [35, 70]. Unlike many other types of cancer cells, however, the KGN cells remain insensitive to these chemotherapeutic agents even after inhibition of NF-κB [35]. In the context of the current study, it is evident that targeting keratin filament expression in GCT and elevating FAS expression offers an alternative, clinical approach to surgical resection or platinum-based chemotherapy. However, therapeutic interventions that impair keratin filament expression/organization and enhance the vulnerability of epithelial cancers to apoptosis might also have the undesirable effect of augmenting tumorigenic potential [33].

Conclusions

In conclusion, the abundance of keratin filaments in KGN cells provides a clinically-relevant mechanism of resistance to FAS-mediated apoptosis by impairing FAS trafficking and possibly activating downstream anti-apoptotic signaling molecules. Here, we provide evidence that decreased expression of FAS at the cell surface, and the presence/activation of labile protein(s) linked to keratin filament expression, provide the means of apoptotic resistance. The existence of keratin filaments in KGN cells and their role in apoptotic resistance provides insight on therapeutic strategies and tumorigenicity of cancers of ovarian origin, specifically GCT and OSE tumors.

Acknowledgements

This work was supported by National Research Initiative Competitive Grant no. 2007-35203-18074 from the USDA National Institute for Food and Agriculture (DHT), and the Olson Center for Women’s Health, Fred and Pamela Buffett Cancer Center at the University of Nebraska Medical Center and the VA Medical Research Program. Partial funding was also provided by the New Hampshire Agricultural Experiment Station. This is Scientific Contribution Number 2644.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare they have no competing interests.

Author’s contributions

SKT, NMS, and XH performed the experiments, conducted the statistical analyses, and assisted in the preparation of the manuscript. SKT, JSD and DHT conceived the study, participated in its design and coordination, and wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
2.
Zurück zum Zitat Schumer ST, Cannistra SA. Granulosa cell tumor of the ovary. J Clin Oncol. 2003;21(6):1180–9.CrossRefPubMed Schumer ST, Cannistra SA. Granulosa cell tumor of the ovary. J Clin Oncol. 2003;21(6):1180–9.CrossRefPubMed
3.
Zurück zum Zitat East N, Alobaid A, Goffin F, Ouallouche K, Gauthier P. Granulosa cell tumour: a recurrence 40 years after initial diagnosis. J Obstet Gynaecol Can. 2005;27(4):363–4.CrossRefPubMed East N, Alobaid A, Goffin F, Ouallouche K, Gauthier P. Granulosa cell tumour: a recurrence 40 years after initial diagnosis. J Obstet Gynaecol Can. 2005;27(4):363–4.CrossRefPubMed
4.
Zurück zum Zitat Hasiakos D, Papakonstantinou K, Karvouni E, Fotiou S. Recurrence of granulosa cell tumor 25 years after initial diagnosis. Report of a case and review of the literature. Eur J Gynaecol Oncol. 2008;29(1):86–8.PubMed Hasiakos D, Papakonstantinou K, Karvouni E, Fotiou S. Recurrence of granulosa cell tumor 25 years after initial diagnosis. Report of a case and review of the literature. Eur J Gynaecol Oncol. 2008;29(1):86–8.PubMed
5.
Zurück zum Zitat Evans 3rd AT, Gaffey TA, Malkasian Jr GD, Annegers JF. Clinicopathologic review of 118 granulosa and 82 theca cell tumors. Obstet Gynecol. 1980;55(2):231–8.PubMed Evans 3rd AT, Gaffey TA, Malkasian Jr GD, Annegers JF. Clinicopathologic review of 118 granulosa and 82 theca cell tumors. Obstet Gynecol. 1980;55(2):231–8.PubMed
6.
Zurück zum Zitat Pectasides D, Papaxoinis G, Fountzilas G, Aravantinos G, Pectasides E, Mouratidou D, et al. Adult granulosa cell tumors of the ovary: a clinicopathological study of 34 patients by the Hellenic Cooperative Oncology Group (HeCOG). Anticancer Res. 2008;28(2B):1421–7.PubMed Pectasides D, Papaxoinis G, Fountzilas G, Aravantinos G, Pectasides E, Mouratidou D, et al. Adult granulosa cell tumors of the ovary: a clinicopathological study of 34 patients by the Hellenic Cooperative Oncology Group (HeCOG). Anticancer Res. 2008;28(2B):1421–7.PubMed
7.
Zurück zum Zitat Amsterdam A, Selvaraj N. Control of differentiation, transformation, and apoptosis in granulosa cells by oncogenes, oncoviruses, and tumor suppressor genes. Endocr Rev. 1997;18(4):435–61.PubMed Amsterdam A, Selvaraj N. Control of differentiation, transformation, and apoptosis in granulosa cells by oncogenes, oncoviruses, and tumor suppressor genes. Endocr Rev. 1997;18(4):435–61.PubMed
8.
Zurück zum Zitat Bridgewater JA, Rustin GJ. Management of non-epithelial ovarian tumours. Oncology. 1999;57(2):89–98.CrossRefPubMed Bridgewater JA, Rustin GJ. Management of non-epithelial ovarian tumours. Oncology. 1999;57(2):89–98.CrossRefPubMed
9.
Zurück zum Zitat Colombo N, Parma G, Zanagnolo V, Insinga A. Management of ovarian stromal cell tumors. J Clin Oncol. 2007;25(20):2944–51.CrossRefPubMed Colombo N, Parma G, Zanagnolo V, Insinga A. Management of ovarian stromal cell tumors. J Clin Oncol. 2007;25(20):2944–51.CrossRefPubMed
10.
Zurück zum Zitat Lee IH, Choi CH, Hong DG, Song JY, Kim YJ, Kim KT, et al. Clinicopathologic characteristics of granulosa cell tumors of the ovary: a multicenter retrospective study. J Gynecol Oncol. 2011;22(3):188–95.PubMedCentralCrossRefPubMed Lee IH, Choi CH, Hong DG, Song JY, Kim YJ, Kim KT, et al. Clinicopathologic characteristics of granulosa cell tumors of the ovary: a multicenter retrospective study. J Gynecol Oncol. 2011;22(3):188–95.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Fuller PJ, Verity K, Shen Y, Mamers P, Jobling T, Burger HG. No evidence of a role for mutations or polymorphisms of the follicle-stimulating hormone receptor in ovarian granulosa cell tumors. J Clin Endocrinol Metab. 1998;83(1):274–9.PubMed Fuller PJ, Verity K, Shen Y, Mamers P, Jobling T, Burger HG. No evidence of a role for mutations or polymorphisms of the follicle-stimulating hormone receptor in ovarian granulosa cell tumors. J Clin Endocrinol Metab. 1998;83(1):274–9.PubMed
12.
Zurück zum Zitat Fuller PJ, Chu S, Jobling T, Mamers P, Healy DL, Burger HG. Inhibin subunit gene expression in ovarian cancer. Gynecol Oncol. 1999;73(2):273–9.CrossRefPubMed Fuller PJ, Chu S, Jobling T, Mamers P, Healy DL, Burger HG. Inhibin subunit gene expression in ovarian cancer. Gynecol Oncol. 1999;73(2):273–9.CrossRefPubMed
13.
Zurück zum Zitat Stouffer RL, Grodin MS, Davis JR, Surwit EA. Investigation of binding sites for follicle-stimulating hormone and chorionic gonadotropin in human ovarian cancers. J Clin Endocrinol Metab. 1984;59(3):441–6.CrossRefPubMed Stouffer RL, Grodin MS, Davis JR, Surwit EA. Investigation of binding sites for follicle-stimulating hormone and chorionic gonadotropin in human ovarian cancers. J Clin Endocrinol Metab. 1984;59(3):441–6.CrossRefPubMed
14.
Zurück zum Zitat Jamieson S, Fuller PJ. Molecular pathogenesis of granulosa cell tumors of the ovary. Endocr Rev. 2012;33(1):109–44.CrossRefPubMed Jamieson S, Fuller PJ. Molecular pathogenesis of granulosa cell tumors of the ovary. Endocr Rev. 2012;33(1):109–44.CrossRefPubMed
15.
Zurück zum Zitat Lee K, Pisarska MD, Ko JJ, Kang Y, Yoon S, Ryou SM, et al. Transcriptional factor FOXL2 interacts with DP103 and induces apoptosis. Biochem Biophys Res Commun. 2005;336(3):876–81.CrossRefPubMed Lee K, Pisarska MD, Ko JJ, Kang Y, Yoon S, Ryou SM, et al. Transcriptional factor FOXL2 interacts with DP103 and induces apoptosis. Biochem Biophys Res Commun. 2005;336(3):876–81.CrossRefPubMed
16.
Zurück zum Zitat Kim JH, Yoon S, Park M, Park HO, Ko JJ, Lee K, et al. Differential apoptotic activities of wild-type FOXL2 and the adult-type granulosa cell tumor-associated mutant FOXL2 (C134W). Oncogene. 2011;30(14):1653–63.CrossRefPubMed Kim JH, Yoon S, Park M, Park HO, Ko JJ, Lee K, et al. Differential apoptotic activities of wild-type FOXL2 and the adult-type granulosa cell tumor-associated mutant FOXL2 (C134W). Oncogene. 2011;30(14):1653–63.CrossRefPubMed
17.
18.
Zurück zum Zitat Kim J, Coffey DM, Ma L, Matzuk MM. The ovary is an alternative site of origin for high-grade serous ovarian cancer in mice. Endocrinology. 2015;156(6):1975–81.CrossRefPubMed Kim J, Coffey DM, Ma L, Matzuk MM. The ovary is an alternative site of origin for high-grade serous ovarian cancer in mice. Endocrinology. 2015;156(6):1975–81.CrossRefPubMed
19.
Zurück zum Zitat Caburet S, Anttonen M, Todeschini AL, Unkila-Kallio L, Mestivier D, Butzow R, et al. Combined comparative genomic hybridization and transcriptomic analyses of ovarian granulosa cell tumors point to novel candidate driver genes. BMC Cancer. 2015;15:251.PubMedCentralCrossRefPubMed Caburet S, Anttonen M, Todeschini AL, Unkila-Kallio L, Mestivier D, Butzow R, et al. Combined comparative genomic hybridization and transcriptomic analyses of ovarian granulosa cell tumors point to novel candidate driver genes. BMC Cancer. 2015;15:251.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Benayoun BA, Anttonen M, L’Hote D, Bailly-Bechet M, Andersson N, Heikinheimo M, et al. Adult ovarian granulosa cell tumor transcriptomics: prevalence of FOXL2 target genes misregulation gives insights into the pathogenic mechanism of the p.Cys134Trp somatic mutation. Oncogene. 2013;32(22):2739–46.CrossRefPubMed Benayoun BA, Anttonen M, L’Hote D, Bailly-Bechet M, Andersson N, Heikinheimo M, et al. Adult ovarian granulosa cell tumor transcriptomics: prevalence of FOXL2 target genes misregulation gives insights into the pathogenic mechanism of the p.Cys134Trp somatic mutation. Oncogene. 2013;32(22):2739–46.CrossRefPubMed
21.
Zurück zum Zitat Park JY, Jin KL, Kim DY, Kim JH, Kim YM, Kim KR, et al. Surgical staging and adjuvant chemotherapy in the management of patients with adult granulosa cell tumors of the ovary. Gynecol Oncol. 2012;125(1):80–6.CrossRefPubMed Park JY, Jin KL, Kim DY, Kim JH, Kim YM, Kim KR, et al. Surgical staging and adjuvant chemotherapy in the management of patients with adult granulosa cell tumors of the ovary. Gynecol Oncol. 2012;125(1):80–6.CrossRefPubMed
22.
Zurück zum Zitat Barak V, Goike H, Panaretakis KW, Einarsson R. Clinical utility of cytokeratins as tumor markers. Clin Biochem. 2004;37(7):529–40.CrossRefPubMed Barak V, Goike H, Panaretakis KW, Einarsson R. Clinical utility of cytokeratins as tumor markers. Clin Biochem. 2004;37(7):529–40.CrossRefPubMed
24.
Zurück zum Zitat Benjamin E, Law S, Bobrow LG. Intermediate filaments cytokeratin and vimentin in ovarian sex cord-stromal tumours with correlative studies in adult and fetal ovaries. J Pathol. 1987;152(4):253–63.CrossRefPubMed Benjamin E, Law S, Bobrow LG. Intermediate filaments cytokeratin and vimentin in ovarian sex cord-stromal tumours with correlative studies in adult and fetal ovaries. J Pathol. 1987;152(4):253–63.CrossRefPubMed
25.
Zurück zum Zitat Kim S, Coulombe PA. Intermediate filament scaffolds fulfill mechanical, organizational, and signaling functions in the cytoplasm. Genes Dev. 2007;21(13):1581–97.CrossRefPubMed Kim S, Coulombe PA. Intermediate filament scaffolds fulfill mechanical, organizational, and signaling functions in the cytoplasm. Genes Dev. 2007;21(13):1581–97.CrossRefPubMed
27.
Zurück zum Zitat Eriksson JE, Dechat T, Grin B, Helfand B, Mendez M, Pallari HM, et al. Introducing intermediate filaments: from discovery to disease. J Clin Invest. 2009;119(7):1763–71.PubMedCentralCrossRefPubMed Eriksson JE, Dechat T, Grin B, Helfand B, Mendez M, Pallari HM, et al. Introducing intermediate filaments: from discovery to disease. J Clin Invest. 2009;119(7):1763–71.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Gilbert S, Loranger A, Daigle N, Marceau N. Simple epithelium keratins 8 and 18 provide resistance to Fas-mediated apoptosis. The protection occurs through a receptor-targeting modulation. J Cell Biol. 2001;154(4):763–73.PubMedCentralCrossRefPubMed Gilbert S, Loranger A, Daigle N, Marceau N. Simple epithelium keratins 8 and 18 provide resistance to Fas-mediated apoptosis. The protection occurs through a receptor-targeting modulation. J Cell Biol. 2001;154(4):763–73.PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Marceau N, Loranger A, Gilbert S, Daigle N, Champetier S. Keratin-mediated resistance to stress and apoptosis in simple epithelial cells in relation to health and disease. Biochem Cell Biol. 2001;79(5):543–55.CrossRefPubMed Marceau N, Loranger A, Gilbert S, Daigle N, Champetier S. Keratin-mediated resistance to stress and apoptosis in simple epithelial cells in relation to health and disease. Biochem Cell Biol. 2001;79(5):543–55.CrossRefPubMed
30.
Zurück zum Zitat Townson DH, Putnam AN, Sullivan BT, Guo L, Irving-Rodgers HF. Expression and distribution of cytokeratin 8/18 intermediate filaments in bovine antral follicles and corpus luteum: an intrinsic mechanism of resistance to apoptosis? Histol Histopathol. 2010;25(7):889–900.PubMed Townson DH, Putnam AN, Sullivan BT, Guo L, Irving-Rodgers HF. Expression and distribution of cytokeratin 8/18 intermediate filaments in bovine antral follicles and corpus luteum: an intrinsic mechanism of resistance to apoptosis? Histol Histopathol. 2010;25(7):889–900.PubMed
31.
Zurück zum Zitat Inada H, Izawa I, Nishizawa M, Fujita E, Kiyono T, Takahashi T, et al. Keratin attenuates tumor necrosis factor-induced cytotoxicity through association with TRADD. J Cell Biol. 2001;155(3):415–26.PubMedCentralCrossRefPubMed Inada H, Izawa I, Nishizawa M, Fujita E, Kiyono T, Takahashi T, et al. Keratin attenuates tumor necrosis factor-induced cytotoxicity through association with TRADD. J Cell Biol. 2001;155(3):415–26.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Hsu H, Shu H-B, Pan M-G, Goeddel DV. TRADD–TRAF2 and TRADD–FADD Interactions Define Two Distinct TNF Receptor 1 Signal Transduction Pathways. Cell. 1996;84(2):299–308.CrossRefPubMed Hsu H, Shu H-B, Pan M-G, Goeddel DV. TRADD–TRAF2 and TRADD–FADD Interactions Define Two Distinct TNF Receptor 1 Signal Transduction Pathways. Cell. 1996;84(2):299–308.CrossRefPubMed
33.
Zurück zum Zitat Fortier AM, Asselin E, Cadrin M. Keratin 8 and 18 loss in epithelial cancer cells increases collective cell migration and cisplatin sensitivity through claudin1 up-regulation. J Biol Chem. 2013;288(16):11555–71.PubMedCentralCrossRefPubMed Fortier AM, Asselin E, Cadrin M. Keratin 8 and 18 loss in epithelial cancer cells increases collective cell migration and cisplatin sensitivity through claudin1 up-regulation. J Biol Chem. 2013;288(16):11555–71.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Nishi Y, Yanase T, Mu Y, Oba K, Ichino I, Saito M, et al. Establishment and characterization of a steroidogenic human granulosa-like tumor cell line, KGN, that expresses functional follicle-stimulating hormone receptor. Endocrinology. 2001;142(1):437–45.CrossRefPubMed Nishi Y, Yanase T, Mu Y, Oba K, Ichino I, Saito M, et al. Establishment and characterization of a steroidogenic human granulosa-like tumor cell line, KGN, that expresses functional follicle-stimulating hormone receptor. Endocrinology. 2001;142(1):437–45.CrossRefPubMed
35.
Zurück zum Zitat Woods DC, White YA, Dau C, Johnson AL. TLR4 activates NF-kappaB in human ovarian granulosa tumor cells. Biochem Biophys Res Commun. 2011;409(4):675–80.PubMedCentralCrossRefPubMed Woods DC, White YA, Dau C, Johnson AL. TLR4 activates NF-kappaB in human ovarian granulosa tumor cells. Biochem Biophys Res Commun. 2011;409(4):675–80.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Matsuda F, Inoue N, Goto Y, Maeda A, Cheng Y, Sakamaki K, et al. cFLIP regulates death receptor-mediated apoptosis in an ovarian granulosa cell line by inhibiting procaspase-8 cleavage. J Reprod Dev. 2008;54(5):314–20.CrossRefPubMed Matsuda F, Inoue N, Goto Y, Maeda A, Cheng Y, Sakamaki K, et al. cFLIP regulates death receptor-mediated apoptosis in an ovarian granulosa cell line by inhibiting procaspase-8 cleavage. J Reprod Dev. 2008;54(5):314–20.CrossRefPubMed
37.
Zurück zum Zitat Quirk SM, Cowan RG, Joshi SG, Henrikson KP. Fas antigen-mediated apoptosis in human granulosa/luteal cells. Biol Reprod. 1995;52(2):279–87.CrossRefPubMed Quirk SM, Cowan RG, Joshi SG, Henrikson KP. Fas antigen-mediated apoptosis in human granulosa/luteal cells. Biol Reprod. 1995;52(2):279–87.CrossRefPubMed
38.
Zurück zum Zitat Duncan A, Forcina J, Birt A, Townson D. Estrous cycle-dependent changes of Fas expression in the bovine corpus luteum: influence of keratin 8/18 intermediate filaments and cytokines. Reprod Biol Endocrinol. 2012;10:90.PubMedCentralCrossRefPubMed Duncan A, Forcina J, Birt A, Townson D. Estrous cycle-dependent changes of Fas expression in the bovine corpus luteum: influence of keratin 8/18 intermediate filaments and cytokines. Reprod Biol Endocrinol. 2012;10:90.PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Caulin C, Ware CF, Magin TM, Oshima RG. Keratin-dependent, epithelial resistance to tumor necrosis factor-induced apoptosis. J Cell Biol. 2000;149(1):17–22.PubMedCentralCrossRefPubMed Caulin C, Ware CF, Magin TM, Oshima RG. Keratin-dependent, epithelial resistance to tumor necrosis factor-induced apoptosis. J Cell Biol. 2000;149(1):17–22.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Coulombe PA, Omary MB. ‘Hard’ and ‘soft’ principles defining the structure, function and regulation of keratin intermediate filaments. Curr Opin Cell Biol. 2002;14(1):110–22.CrossRefPubMed Coulombe PA, Omary MB. ‘Hard’ and ‘soft’ principles defining the structure, function and regulation of keratin intermediate filaments. Curr Opin Cell Biol. 2002;14(1):110–22.CrossRefPubMed
41.
Zurück zum Zitat Czernobilsky B, Moll R, Levy R, Franke WW. Co-expression of cytokeratin and vimentin filaments in mesothelial, granulosa and rete ovarii cells of the human ovary. Eur J Cell Biol. 1985;37:175–90.PubMed Czernobilsky B, Moll R, Levy R, Franke WW. Co-expression of cytokeratin and vimentin filaments in mesothelial, granulosa and rete ovarii cells of the human ovary. Eur J Cell Biol. 1985;37:175–90.PubMed
42.
Zurück zum Zitat Wendl J, Ebach K, Rodler D, Kenngott RA. Immunocytochemical localization of cytoplasmic and nuclear intermediate filaments in the bovine ovary during folliculogenesis. Anat Histol Embryol. 2012;41(3):190–201.CrossRefPubMed Wendl J, Ebach K, Rodler D, Kenngott RA. Immunocytochemical localization of cytoplasmic and nuclear intermediate filaments in the bovine ovary during folliculogenesis. Anat Histol Embryol. 2012;41(3):190–201.CrossRefPubMed
43.
Zurück zum Zitat Ricken AM, Spanel-Borowski K, Saxer M, Huber PR. Cytokeratin expression in bovine corpora lutea. Histochem Cell Biol. 1995;103(5):345–54.CrossRefPubMed Ricken AM, Spanel-Borowski K, Saxer M, Huber PR. Cytokeratin expression in bovine corpora lutea. Histochem Cell Biol. 1995;103(5):345–54.CrossRefPubMed
44.
Zurück zum Zitat Santini D, Ceccarelli C, Mazzoleni G, Pasquinelli G, Jasonni VM, Martinelli GN. Demonstration of cytokeratin intermediate filaments in oocytes of the developing and adult human ovary. Histochemistry. 1993;99(4):311–9.CrossRefPubMed Santini D, Ceccarelli C, Mazzoleni G, Pasquinelli G, Jasonni VM, Martinelli GN. Demonstration of cytokeratin intermediate filaments in oocytes of the developing and adult human ovary. Histochemistry. 1993;99(4):311–9.CrossRefPubMed
45.
Zurück zum Zitat Sutovsky P, Flechon JE, Pavlok A. Microfilaments, microtubules and intermediate filaments fulfil differential roles during gonadotropin-induced expansion of bovine cumulus oophorus. Reprod Nutr Dev. 1994;34(5):415–25.CrossRefPubMed Sutovsky P, Flechon JE, Pavlok A. Microfilaments, microtubules and intermediate filaments fulfil differential roles during gonadotropin-induced expansion of bovine cumulus oophorus. Reprod Nutr Dev. 1994;34(5):415–25.CrossRefPubMed
46.
Zurück zum Zitat van den Hurk R, Dijkstra G, van Mil FN, Hulshof SC, van den Ingh TS. Distribution of the intermediate filament proteins vimentin, keratin, and desmin in the bovine ovary. Mol Reprod Dev. 1995;41(4):459–67.CrossRefPubMed van den Hurk R, Dijkstra G, van Mil FN, Hulshof SC, van den Ingh TS. Distribution of the intermediate filament proteins vimentin, keratin, and desmin in the bovine ovary. Mol Reprod Dev. 1995;41(4):459–67.CrossRefPubMed
48.
49.
Zurück zum Zitat Vickers SL, Cowan RG, Harman RM, Porter DA, Quirk SM. Expression and activity of the Fas antigen in bovine ovarian follicle cells. Biol Reprod. 2000;62(1):54–61.CrossRefPubMed Vickers SL, Cowan RG, Harman RM, Porter DA, Quirk SM. Expression and activity of the Fas antigen in bovine ovarian follicle cells. Biol Reprod. 2000;62(1):54–61.CrossRefPubMed
50.
Zurück zum Zitat Hu S, Vincenz C, Ni J, Gentz R, Dixit VM. I-FLICE, a novel inhibitor of tumor necrosis factor receptor-1- and CD-95-induced apoptosis. J Biol Chem. 1997;272(28):17255–7.CrossRefPubMed Hu S, Vincenz C, Ni J, Gentz R, Dixit VM. I-FLICE, a novel inhibitor of tumor necrosis factor receptor-1- and CD-95-induced apoptosis. J Biol Chem. 1997;272(28):17255–7.CrossRefPubMed
51.
Zurück zum Zitat Matsuda-Minehata F, Goto Y, Inoue N, Sakamaki K, Chedrese PJ, Manabe N. Anti-apoptotic activity of porcine cFLIP in ovarian granulosa cell lines. Mol Reprod Dev. 2007;74(9):1165–70.CrossRefPubMed Matsuda-Minehata F, Goto Y, Inoue N, Sakamaki K, Chedrese PJ, Manabe N. Anti-apoptotic activity of porcine cFLIP in ovarian granulosa cell lines. Mol Reprod Dev. 2007;74(9):1165–70.CrossRefPubMed
52.
Zurück zum Zitat Kreuz S, Siegmund D, Scheurich P, Wajant H. NF-kappaB inducers upregulate cFLIP, a cycloheximide-sensitive inhibitor of death receptor signaling. Mol Cell Biol. 2001;21(12):3964–73.PubMedCentralCrossRefPubMed Kreuz S, Siegmund D, Scheurich P, Wajant H. NF-kappaB inducers upregulate cFLIP, a cycloheximide-sensitive inhibitor of death receptor signaling. Mol Cell Biol. 2001;21(12):3964–73.PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Irmler M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner V, et al. Inhibition of death receptor signals by cellular FLIP. Nature. 1997;388(6638):190–5.CrossRefPubMed Irmler M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner V, et al. Inhibition of death receptor signals by cellular FLIP. Nature. 1997;388(6638):190–5.CrossRefPubMed
54.
Zurück zum Zitat Scaffidi C, Schmitz I, Krammer PH, Peter ME. The role of c-FLIP in modulation of CD95-induced apoptosis. J Biol Chem. 1999;274(3):1541–8.CrossRefPubMed Scaffidi C, Schmitz I, Krammer PH, Peter ME. The role of c-FLIP in modulation of CD95-induced apoptosis. J Biol Chem. 1999;274(3):1541–8.CrossRefPubMed
55.
Zurück zum Zitat Tschopp J, Irmler M, Thome M. Inhibition of fas death signals by FLIPs. Curr Opin Immunol. 1998;10(5):552–8.CrossRefPubMed Tschopp J, Irmler M, Thome M. Inhibition of fas death signals by FLIPs. Curr Opin Immunol. 1998;10(5):552–8.CrossRefPubMed
56.
Zurück zum Zitat Steinmetz R, Wagoner HA, Zeng P, Hammond JR, Hannon TS, Meyers JL, et al. Mechanisms regulating the constitutive activation of the extracellular signal-regulated kinase (ERK) signaling pathway in ovarian cancer and the effect of ribonucleic acid interference for ERK1/2 on cancer cell proliferation. Mol Endocrinol. 2004;18(10):2570–82.CrossRefPubMed Steinmetz R, Wagoner HA, Zeng P, Hammond JR, Hannon TS, Meyers JL, et al. Mechanisms regulating the constitutive activation of the extracellular signal-regulated kinase (ERK) signaling pathway in ovarian cancer and the effect of ribonucleic acid interference for ERK1/2 on cancer cell proliferation. Mol Endocrinol. 2004;18(10):2570–82.CrossRefPubMed
57.
Zurück zum Zitat Wang C, Lv X, Jiang C, Cordes CM, Fu L, Lele SM, et al. Transforming growth factor alpha (TGFalpha) regulates granulosa cell tumor (GCT) cell proliferation and migration through activation of multiple pathways. PLoS ONE. 2012;7(11):e48299.PubMedCentralCrossRefPubMed Wang C, Lv X, Jiang C, Cordes CM, Fu L, Lele SM, et al. Transforming growth factor alpha (TGFalpha) regulates granulosa cell tumor (GCT) cell proliferation and migration through activation of multiple pathways. PLoS ONE. 2012;7(11):e48299.PubMedCentralCrossRefPubMed
58.
Zurück zum Zitat Rico C, Lague MN, Lefevre P, Tsoi M, Dodelet-Devillers A, Kumar V, et al. Pharmacological targeting of mammalian target of rapamycin inhibits ovarian granulosa cell tumor growth. Carcinogenesis. 2012;33(11):2283–92.CrossRefPubMed Rico C, Lague MN, Lefevre P, Tsoi M, Dodelet-Devillers A, Kumar V, et al. Pharmacological targeting of mammalian target of rapamycin inhibits ovarian granulosa cell tumor growth. Carcinogenesis. 2012;33(11):2283–92.CrossRefPubMed
59.
Zurück zum Zitat Hu CL, Cowan RG, Harman RM, Quirk SM. Cell cycle progression and activation of Akt kinase are required for insulin-like growth factor I-mediated suppression of apoptosis in granulosa cells. Mol Endocrinol. 2004;18(2):326–38.CrossRefPubMed Hu CL, Cowan RG, Harman RM, Quirk SM. Cell cycle progression and activation of Akt kinase are required for insulin-like growth factor I-mediated suppression of apoptosis in granulosa cells. Mol Endocrinol. 2004;18(2):326–38.CrossRefPubMed
60.
Zurück zum Zitat Fluhr H, Spratte J, Bredow M, Heidrich S, Zygmunt M. Constitutive activity of Erk1/2 and NF-kappaB protects human endometrial stromal cells from death receptor-mediated apoptosis. Reprod Biol. 2013;13(2):113–21.CrossRefPubMed Fluhr H, Spratte J, Bredow M, Heidrich S, Zygmunt M. Constitutive activity of Erk1/2 and NF-kappaB protects human endometrial stromal cells from death receptor-mediated apoptosis. Reprod Biol. 2013;13(2):113–21.CrossRefPubMed
61.
Zurück zum Zitat Gibson S, Tu S, Oyer R, Anderson SM, Johnson GL. Epidermal growth factor protects epithelial cells against Fas-induced apoptosis. Requirement for Akt activation. J Biol Chem. 1999;274(25):17612–8.CrossRefPubMed Gibson S, Tu S, Oyer R, Anderson SM, Johnson GL. Epidermal growth factor protects epithelial cells against Fas-induced apoptosis. Requirement for Akt activation. J Biol Chem. 1999;274(25):17612–8.CrossRefPubMed
62.
Zurück zum Zitat Toivola DM, Nieminen MI, Hesse M, He T, Baribault H, Magin TM, et al. Disturbances in hepatic cell-cycle regulation in mice with assembly-deficient keratins 8/18. Hepatology. 2001;34(6):1174–83.CrossRefPubMed Toivola DM, Nieminen MI, Hesse M, He T, Baribault H, Magin TM, et al. Disturbances in hepatic cell-cycle regulation in mice with assembly-deficient keratins 8/18. Hepatology. 2001;34(6):1174–83.CrossRefPubMed
63.
Zurück zum Zitat Galarneau L, Loranger A, Gilbert S, Marceau N. Keratins modulate hepatic cell adhesion, size and G1/S transition. Exp Cell Res. 2007;313(1):179–94.CrossRefPubMed Galarneau L, Loranger A, Gilbert S, Marceau N. Keratins modulate hepatic cell adhesion, size and G1/S transition. Exp Cell Res. 2007;313(1):179–94.CrossRefPubMed
64.
Zurück zum Zitat Oshima RG. Apoptosis and keratin intermediate filaments. Cell Death Differ.2002;9(5):486-92. Oshima RG. Apoptosis and keratin intermediate filaments. Cell Death Differ.2002;9(5):486-92.
65.
Zurück zum Zitat Sullivan BT, Cherry JA, Sakamoto H, Henkes LE, Townson DH, Rueda BR. Cytokeratin 18 expression inhibits cytokine-induced death of cervical cancer cells. Int J Gynecol Cancer. 2010;20(9):1474–81.PubMed Sullivan BT, Cherry JA, Sakamoto H, Henkes LE, Townson DH, Rueda BR. Cytokeratin 18 expression inhibits cytokine-induced death of cervical cancer cells. Int J Gynecol Cancer. 2010;20(9):1474–81.PubMed
66.
Zurück zum Zitat Wu MS, Li CH, Ruppert JG, Chang CC. Cytokeratin 8-MHC class I interactions: a potential novel immune escape phenotype by a lymph node metastatic carcinoma cell line. Biochem Biophys Res Commun. 2013;441(3):618–23.CrossRefPubMed Wu MS, Li CH, Ruppert JG, Chang CC. Cytokeratin 8-MHC class I interactions: a potential novel immune escape phenotype by a lymph node metastatic carcinoma cell line. Biochem Biophys Res Commun. 2013;441(3):618–23.CrossRefPubMed
67.
Zurück zum Zitat Mackinder MA, Evans CA, Chowdry J, Staton CA, Corfe BM. Alteration in composition of keratin intermediate filaments in a model of breast cancer progression and the potential to reverse hallmarks of metastasis. Cancer biomarkers : section A of Disease markers. 2012;12(2):49–64. Mackinder MA, Evans CA, Chowdry J, Staton CA, Corfe BM. Alteration in composition of keratin intermediate filaments in a model of breast cancer progression and the potential to reverse hallmarks of metastasis. Cancer biomarkers : section A of Disease markers. 2012;12(2):49–64.
68.
Zurück zum Zitat Holdenrieder S, Stieber P, Liska V, Treska V, Topolcan O, Dreslerova J, et al. Cytokeratin serum biomarkers in patients with colorectal cancer. Anticancer Res. 2012;32(5):1971–6.PubMed Holdenrieder S, Stieber P, Liska V, Treska V, Topolcan O, Dreslerova J, et al. Cytokeratin serum biomarkers in patients with colorectal cancer. Anticancer Res. 2012;32(5):1971–6.PubMed
69.
Zurück zum Zitat Iyer SV, Dange PP, Alam H, Sawant SS, Ingle AD, Borges AM, et al. Understanding the role of keratins 8 and 18 in neoplastic potential of breast cancer derived cell lines. PLoS ONE. 2013;8(1):e53532.PubMedCentralCrossRefPubMed Iyer SV, Dange PP, Alam H, Sawant SS, Ingle AD, Borges AM, et al. Understanding the role of keratins 8 and 18 in neoplastic potential of breast cancer derived cell lines. PLoS ONE. 2013;8(1):e53532.PubMedCentralCrossRefPubMed
70.
Zurück zum Zitat Woods DC, Alvarez C, Johnson AL. Cisplatin-mediated sensitivity to TRAIL-induced cell death in human granulosa tumor cells. Gynecol Oncol. 2008;108(3):632–40.CrossRefPubMed Woods DC, Alvarez C, Johnson AL. Cisplatin-mediated sensitivity to TRAIL-induced cell death in human granulosa tumor cells. Gynecol Oncol. 2008;108(3):632–40.CrossRefPubMed
Metadaten
Titel
Molecular manipulation of keratin 8/18 intermediate filaments: modulators of FAS-mediated death signaling in human ovarian granulosa tumor cells
Publikationsdatum
01.12.2016
Erschienen in
Journal of Ovarian Research / Ausgabe 1/2016
Elektronische ISSN: 1757-2215
DOI
https://doi.org/10.1186/s13048-016-0217-z

Weitere Artikel der Ausgabe 1/2016

Journal of Ovarian Research 1/2016 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.