Skip to main content
Erschienen in: BMC Cancer 1/2017

Open Access 01.12.2017 | Research article

The tumor suppressor RhoBTB1 controls Golgi integrity and breast cancer cell invasion through METTL7B

verfasst von: Caroline M. McKinnon, Harry Mellor

Erschienen in: BMC Cancer | Ausgabe 1/2017

Abstract

Background

RhoBTB1 and 2 are atypical members of the Rho GTPase family of signaling proteins. Unlike other Rho GTPases, RhoBTB1 and 2 undergo silencing or mutation in a wide range of epithelial cancers; however, little is known about the consequences of this loss of function.

Methods

We analyzed transcriptome data to identify transcriptional targets of RhoBTB2. We verified these using Q-PCR and then used gene silencing and cell imaging to determine the cellular function of these targets downstream of RhoBTB signaling.

Results

RhoBTB1 and 2 regulate the expression of the methyltransferases METTL7B and METTL7A, respectively. RhoBTB1 regulates the integrity of the Golgi complex through METTL7B. RhoBTB1 is required for expression of METTL7B and silencing of either protein leads to fragmentation of the Golgi. Loss of RhoBTB1 expression is linked to Golgi fragmentation in breast cancer cells. Restoration of normal RhoBTB1 expression rescues Golgi morphology and dramatically inhibits breast cancer cell invasion.

Conclusion

Loss of RhoBTB1 expression in breast cancer cells leads to Golgi fragmentation and hence loss of normal polarity.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12885-017-3138-3) contains supplementary material, which is available to authorized users.
Abkürzungen
BTB
Broad-complex, tramtrack and bric a brac
ER
Endoplasmic reticulum
HNSC
Head and neck squamous cell
METTL
Methyltransferase like
RT-PCR
Real-time PCR.

Background

The Rho GTPase family of signaling proteins are master regulators of cell shape and cell migration. They do this directly through dynamic regulation of the actin cytoskeleton; however, they also have diverse additional cellular roles that contribute to this, including the control of membrane trafficking, cell polarity and gene expression [1]. The roles of Rho GTPases in cell migration make them important signaling proteins in cancer. While Rho GTPases are generally not direct targets of mutation in cancer, their signaling pathways are frequently deregulated, promoting the switch to cancer cell invasion and metastasis [2, 3].
The human Rho GTPase family contains 20 members, of which RhoA, Rac1 and Cdc42 are the best characterized [4]. These are small, globular proteins whose activity is controlled by binding of GTP, which switches them into their active conformation. The Rho family also contains two atypical members – RhoBTB1 and 2. These are larger, multimodular Rho GTPases that have a conserved N-terminal Rho GTPase domain, but also two copies of the BTB (Broad-Complex, Tramtrack and Bric a brac) domain and a carboxyl terminal BACK (BTB and C-terminal Kelch) domain [5, 6]. Intriguingly, both genes undergo silencing or mutation in human cancer. Hamaguchi and colleagues identified RhoBTB2 in a representational difference analysis screen for novel tumor suppressor genes in breast cancer, and gave it the alternative name DBC2 (deleted in breast cancer 2). The RhoBTB2/DBC2 gene undergoes homologous deletion in a relatively small number of breast tumor samples; however, RhoBTB2 expression is silenced at high frequency (approximately 50%) in breast and lung tumors [7]. Subsequent studies have reported the silencing of RhoBTB2 expression in a wide range of human tumors, as well as sporadic point mutations of the RhoBTB2 coding region and promoter [811]. RhoBTB1 is 73% identical to RhoBTB2 at the protein level. Far less is known about its cellular functions; however, recent studies have shown that it is also downregulated in human cancers. It is subject to loss of heterozygosity at high frequency in head and neck squamous cell (HNSC) carcinomas [12] and its expression is silenced in colon cancer through the actions of the microRNA miR-31 [13].
Unlike the majority of members of the Rho GTPase family, RhoBTB1 and 2 do not regulate the actin cytoskeleton directly [14]. Many proteins with BTB domains function as transcription regulators [15] and in our previous studies we showed that this is also the case for RhoBTB2 [16]. To determine transcription targets of RhoBTB2, we silenced its expression in primary lung epithelial cells and then performed whole-genome microarray analysis of gene expression. This allowed us to identify the chemokine CXCL14 as a target of RhoBTB2 regulation [16]. CXCL14 expression is downregulated in a high percentage of carcinomas, and especially in HNSC carcinomas where its loss is correlated with poor prognosis. Importantly, we found that loss of RhoBTB2 expression is correlated with loss of CXCL14 expression in HNSC cancer cell lines, and that expression of the chemokine is rescued by re-expression of RhoBTB2 [16].
CXCL14 was the most significant hit in the RhoBTB2 microarray screen; however, several other genes also showed reduced expression upon RhoBTB2 silencing. One of these was METTL7A, a poorly-characterized methyltransferase enzyme. In this study, we investigate the regulation of the METTL7 enzymes by RhoBTB proteins and uncover a pathway controlling Golgi integrity in mammary epithelial cells.

Methods

Materials

Full details of antibodies, oligonucleotides and plasmids used in this study are given in Additional file 1.

Cell culture and transfection

HeLa, HEK293T, MDA-MB-231, MCF7 and T47D cells were cultured in DMEM containing 10% heat-inactivated fetal bovine serum. HMT-S1 and MCF10A cells were cultured as previously described [17, 18]. HeLa cells were transfected with plasmids and siRNA oligonucleotides using calcium phosphate [16].

Real-time PCR

RNA was isolated from cells using the TRIzol extraction method (Invitrogen) and 40 μg of purified RNA used for reverse transcription using Omniscript RTase (Qiagen) for 1 h at 37 °C. cDNAs were then subjected to real-time PCR using DyNAmo Flash SYBR Green (Finnzymes). Amplification was performed using an Opticon 2 thermocycler (MJ Research) and data was analyzed using the comparative Ct method.

Immunofluorescence microscopy

Cells were fixed for 15 min in 4% paraformaldehyde in PBS and then permeabilized in 0.2% Triton X-100 in PBS for 5 min. Cells were then incubated with 0.1% sodium borohydride for 10 min. Primary antibodies were incubated with cells in 1% BSA for 1 h followed by secondary antibodies for 45 min. The cells were stained with 2 μg/ml DAPI for 10 min and mounted over MOWIOL 4–88 (Calbiochem) containing 0.6% 1,4-diazabicyclo-(2.2.2) octane as an anti-photobleaching agent. Confocal microscopy was performed using a Leica TCS-NT confocal laser-scanning microscope with an attached Leica DMRBE upright epifluorescence microscope under a PlanApo x63/1.32 oil-immersion objective. A series of images were taken at 0.5 μm intervals through the Z-plane of the cells and processed to form a projected image.

Analysis of Golgi fragmentation

Fragmentation of the Golgi ribbon was scored blind in cells stained for the Golgi marker giantin. Fifty cells were scored from each condition to give the percentage of cells with a fragmented Golgi. Data from three independent experiments were processed to give the mean.

Re-expression of RhoBTB1

Expression of RhoBTB1 was restored in T47D cells by stable integration of a lentiviral RhoBTB1 construct. mCherry-tagged RhoBTB1 was subcloned into pHR’SIN-cPPT-SEW [19]. Virus was generated by transfection of HEK293T cells as described [19]. Briefly, 40 μg of RhoBTB1 vector was mixed with 10 μg of envelope plasmid pMDG, 30 μg of packaging plasmid psPAX2 and 1 μl of 1 mM polyethylenimine (Sigma) in OptiMEM (Invitrogen). This transfection mixture was replaced 4 h later with 15 ml of normal culture medium. The cells were then incubated for 48 h to allow virus production. After the incubation, the medium was removed and centrifuged for 10 min at 2,600 x g. The supernatant was then passed through a 0.45 μm filter and this filtrate was used as the virus stock. T47D cells were transduced with virus stock by overnight incubation.

Cell migration assays

T47D cells were grown to confluence in chamber slides (Ibidi). The confluent monolayer was scratched with a sterile pipette tip and migration was followed by brightfield imaging at 37 °C for 14 h using a Leica AF6000 live cell imaging workstation and x40 objective. Multipoint revisiting allowed for parallel imaging of samples. Average cell velocity was calculated using ImageJ (NIH). For quantification of Golgi polarization, cells were fixed 1 h after making the scratch and stained for giantin. Cell polarization was assessed by dividing cells into quadrants centered on the nucleus. Cells were judged to have polarized if the majority of their Golgi apparatus resided within the quadrant facing the scratch edge.

Cell invasion assays

Invasion of T47D cells was quantified using BD BioCoat Matrigel Invasion Chambers (BD Biosciences) according to the manufacturer’s protocol. Briefly, T47D cells in serum-free medium were plated at 1 × 105 cells per chamber. Medium containing 10% fetal bovine serum was used as the chemoattractant in the lower chamber. After 24 h, non-invaded cells were removed from the upper chamber using a cotton swab. Cells that invaded through the Matrigel to the bottom of the insert were fixed and stained by incubation with Diff-Quick (BD Biosciences) for 10 min. These stained cells were washed with PBS, air-dried and counted.

Statistical analysis

Analysis of statistical significance was performed using GraphPad Prism software. Comparison of two sample experiments was by paired, two-tailed Student’s t-test. Analysis of multiple samples was made using one-way ANOVA. For comparisons to a control, Dunnett’s post hoc test was used, whereas comparisons between all samples employed Tukey’s post hoc test.

Results

RhoBTBs control the expression of METTL7 isoforms

Our previous microarray screen identified METTL7A as a potential target of RhoBTB2-mediated transcriptional regulation [16]. To validate this, we used two independent siRNAs to silence RhoBTB2 in HeLa cells and measured METTL7A expression by RT-PCR. We also examined the expression of METTL7B, a closely-related protein that shares 59% sequence identity with METTL7A. Silencing of RhoBTB2 reduced the expression of METTL7A by approximately 30% (Fig. 1a). Previously, we showed that both RhoBTB1 and RhoBTB2 are required for expression of CXCL14 [16]; however, silencing of RhoBTB1 had no effect on METTLA expression (Fig. 1b). Interestingly, RhoBTB1 instead regulated the expression of METTL7B (Fig. 1b). We conclude that the two human METTL7 isoforms are differentially regulated by these two RhoBTBs.

RhoBTB1 regulates Golgi integrity

METTL7B was originally identified as a Golgi-associated methyltransferase of unknown function [20]. Previous work has shown that inhibition of cellular methylation leads to fragmentation of Golgi, although the identity of the methyltransferase involved is unknown [21]. We hypothesized that the RhoBTBs might act through METTL7 proteins to control some aspect of Golgi function. To examine this, we silenced RhoBTB1 and RhoBTB2 expression and examined the basic morphology of the Golgi by staining cells for the well-characterized Golgi marker, giantin. In control cells, the Golgi was present as a characteristic ribbon situated next to the nucleus. Silencing of RhoBTB2 had a negligible effect on Golgi morphology; however, silencing of RhoBTB1 caused profound fragmentation of the Golgi (Fig. 2a, b).

RhoBTB1 regulates Golgi integrity through METTL7B

We designed specific siRNAs to each METTL7 isoform to allow us to investigate their role in the regulation of Golgi integrity (Fig. 3a, b). Interestingly, silencing of either isoform caused significant fragmentation of the Golgi (Fig. 3c, d). This raised the possibility that downregulation of METTL7B might underlie the effects of RhoBTB1 depletion on Golgi integrity. To test this, we silenced RhoBTB1 and then restored METTL7B expression by transfection. This re-expression of METTL7B was able to significantly rescue Golgi integrity (Fig. 3e, f). Intriguingly, expression of METTL7A also promoted recue of normal Golgi morphology (Fig. 3e, f). We conclude that the effects of RhoBTB1 depletion on Golgi integrity are mediated through the consequent reduced expression of METTL7B, but that METTL7A overexpression can substitute for this activity.
METTL7B has been reported as being localized to the Golgi [20], whereas METTL7A (a.k.a. AAM-B) has previously been shown to localize to the endoplasmic reticulum (ER) and to lipid droplets [22]. It was unclear why both isoforms should affect Golgi morphology, given their different locations. We examined the cellular localization of both proteins. In contrast to the previous report [20], we found that both METTL7s localized to the ER in HeLa cells, with no apparent Golgi localization (Fig. 4a, b). We conclude that the relevant target(s) for METTL7 action is likely to be an ER resident protein, or a protein that actively shuttles between the two compartments.

RhoBTB1 is expression is downregulated in breast cancer cell lines

Many studies have reported downregulation of RhoBTB2 expression in breast cancer, most commonly due to methylation of the RhoBTB2 promoter [7, 10, 11, 23, 24]. We examined the relative expression of RhoBTB1 and RhoBTB2 in a selection of human breast cancer cell lines (MDA-MB-231, T47D, MCF7) in comparison to two well-characterized normal human mammary epithelial cell lines – MCF10A [25] and HMT-3522 S1 [26]. All three breast cancer cell lines had reduced RhoBTB1 expression (Fig. 5a). T47D and MDA-MB-231 cells also showed reduced expression of RhoBTB2, whereas expression was normal in MCF7 cells (Fig. 5b). We examined the expression of METTL7A and METTL7B in the same cell lines. Interestingly, both isoforms showed reduced expression in the breast cancer cell lines, with T47D cells having the lowest overall expression of METTL7 isoforms (Fig. 5c, d). T47D cells have previously been shown to undergone loss of RhoBTB2 expression [7]. They also had the lowest expression of RhoBTB1 (Fig. 5a). Intriguingly, whereas the normal mammary cell lines had a compact Golgi ribbon, T47D cells showed extreme fragmentation of the Golgi (Figs. 5e, 6b), raising the possibility of a functional link between loss of RhoBTB1 expression and Golgi morphology in this breast cancer line.

Restoration of RhoBTB1 expression restores normal Golgi morphology to T47D cells and inhibits invasion

To explore the effect of loss of RhoBTB1 expression on Golgi integrity in T47D cells, we restored RhoBTB1 expression using lentiviral transduction. The resultant stable cell line had near normal levels of RhoBTB1, but still had dramatically lower levels of RhoBTB2 (Fig. 6a). Restoration of RhoBTB1 expression led to a significant rescue of Golgi integrity (Fig. 6b).
Fragmentation of the Golgi has little effect on the rate of protein secretion in cells [27]. In keeping with this, we saw no apparent change in the rate of trafficking of VSV-G through the secretory pathway in cells depleted of RhoBTB1 (data not shown). One proposed function of the Golgi ribbon structure is to facilitate the cellular localization of this organelle. Polarized epithelial cells orientate their Golgi towards the apical surface of the cell, and this is important for polarized secretion and for maintenance of the apical-basolateral axis [28]. In migrating cells, the Golgi is polarized towards the leading edge of the cell in the majority of cell types and this reinforces directional migration [29]. Dysregulation of cell polarity pathways is an important component in the conversion of cancer cells into a migratory, invasive phenotype. We examined whether restoration of RhoBTB1 expression and normalization of Golgi morphology would affect these processes in T47D breast cancer cells. We used a scratch migration assay to measure Golgi polarization and the directional migration of cells. T47D cells failed to polarize their Golgi effectively in the scratch assay; whereas restoration of RhoBTB1 expression resulted in strong polarization of the Golgi towards the migration front (Fig. 6d). Restoration of RhoBTB1 expression had no effect on T47D cell migration, however, suggesting that the Golgi fragmentation observed in T47D cells does not affect their migration in 2D (Fig. 6e). We next examined the invasive capacity of these cells. A critical early stage in breast cancer progression is breakdown of the highly polarized organization of cells in a mammary duct and their invasion through the basement lamina and into the surrounding stroma. We measured the ability of T47D breast cancer cells to invade through a 3D layer of extracellular matrix that mimics the composition of the basement lamina [30]. Restoration of RhoBTB1 expression dramatically inhibited cell invasion (Fig. 6f). We conclude that loss of RhoBTB1 expression in T47D breast cancer cells contributes to a pro-invasion phenotype. Our hypothesis is that this is a result of reduced METTL7B expression, leading to Golgi fragmentation and a breakdown of normal cell polarity.

Discussion

RhoBTB proteins are highly unusual members of the RhoGTPase family. Their multidomain structure makes them atypical; however, they are present in a wide range of organisms, including the social amoeba Dictyostelium discoideum, where they were first identified [31]. We still know very little about the cellular functions of RhoBTB1 and 2. Most progress has come from investigating the function of their twin BTB domains [6]. In many proteins, the BTB domain functions to recruit the Cul3 ubiquitin ligase [32]. RhoBTB2 can bind Cul3 through its first BTB domain [33, 34]. RhoBTB2 itself is a target of ubiquitylation by this complex, leading to its subsequent degradation by the proteasome [34]. This raises the possibility of a role for the RhoBTB2/Cul3 complex in regulating the turnover of other cellular proteins, although no targets of RhoBTB2/Cul3 mediated ubiquitylation have yet been identified.
BTB domains are also frequently found in transcriptional regulators, where they have a number of roles, including the recruitment of transcriptional co-repressors [35]. Previously, we showed that RhoBTB2 controls the expression of the CXCL14 chemokine in epithelial cells [16]. Here were show that the METTL7 enzymes are targets of transcriptional control by RhoBTB1 and 2. CXCL14 expression requires both RhoBTB1 and 2 [16], whereas the two RhoBTBs had specific roles in regulating the METTL7 enzymes, with RhoBTB2 regulating expression of METTL7A and RhoBTB1 regulating METTL7B. The RhoBTBs dimerize through their BTB domains, and can form both homo- and heterodimers [33]. It seems likely these different dimer pairs have different transcriptional targets. This becomes important when considering the effects of the downregulation of RhoBTB1 and 2 in cancer cells, where loss of one isoform could potentially increase homodimer concentration of the other, in addition to the simpler effects of reducing its own concentration.
We show here that RhoBTB1 regulates Golgi integrity. Interestingly, the third RhoBTB family member, RhoBTB3, has also been shown to regulate Golgi function. RhoBTB3 is only weakly-related to RhoBTB1 and 2, but shares a similar domain organization [6]. RhoBTB3 is a Golgi-localized protein and binds to directly to the secretory pathway regulator Rab9 to mediate trafficking from the endoplasmic reticulum to the Golgi [36]. RhoBTB3 silencing has been reported to lead to enlargement of the Golgi [36] and in a later study to cause Golgi fragmentation[37] The mechanism by which RhoBTB3 affects Golgi integrity has not been reported. We show here that RhoBTB1 controls Golgi integrity through regulation of the expression of METTL7B. The downstream targets of the poorly-characterized METTL7 enzymes are unknown. Both contain an N-terminal transmembrane domain and a C-terminal methyltransferase domain, predicted to lie on the cytoplasmic face. Interestingly, METTL7B undergoes arginine dimethylation [20], raising the possibility of automethylation. Depletion of either METTL7A or B caused Golgi fragmentation (Fig. 3c) and METTL7A overexpression was able to rescue the effects of depletion of METTL7B (Fig. 3e). This suggests that the two proteins have overlapping functions in regulating Golgi integrity. Silencing of RhoBTB2 did not cause significant Golgi fragmentation (Fig. 2b), despite a reduction in METTL7A expression (Fig. 1a). The effects of RhoBTB2 on METTL7A are less pronounced than those of RhoBTB1 on METTL7B, at least in HeLa cells (Fig. 1). It is possible that RhoBTB2 might affect Golgi integrity in other cell types, perhaps where METTL7A is the more dominant isoform.
In vertebrates, the Golgi apparatus is organized into a compact ribbon structure, typically located next to the nucleus [38]. This organization depends on the constant action of the motor protein dynein and on the functions of Golgi structural proteins [29, 38]. Surprisingly, Golgi ribbon structure is not requisite for the functioning of the secretory pathway. In plants, individual Golgi stacks are scattered throughout the cytosol, and in the yeast Saccharomyces cerevisiae, the Golgi is composed of dispersed, isolated cisternae [38, 39]. In vertebrate cells, treatments that fragment the Golgi do not affect the rate of protein secretion [40, 41]. Instead, the organized Golgi ribbon structure seems to be important for vertebrate cell polarization. In polarized epithelial cells, the Golgi is orientated towards the apical surface [28]. In most migrating vertebrate cells, the Golgi is orientated towards the leading edge [29, 42]. These observations suggest that a polarized Golgi ribbon may facilitate polarized secretion. In polarized epithelial cells, this would support apical/basolateral polarity, and in migrating cells, it would contribute to directional movement. In keeping with this, recent work supports a role for the Golgi in the polarized delivery of active Cdc42 to the leading edge of migrating cells [43]. Fragmentation of the Golgi has previously been shown to inhibit the migration of HeLa cells [27]. Here we find that the loss of expression of RhoBTB1 in T47D breast cancer cells is linked to fragmentation of the Golgi. This can be rescued by re-expression of RhoBTB1; however, this has no effect on migration of these cells in 2D (Fig. 6e). Instead, we found that re-expression of RhoBTB1 in T47D breast cancer cells strongly inhibited their invasive capacity in 3D (Fig. 6f). The initial stages of breast cancer progression involve loss of apical-basolateral polarity and invasion through the basement lamina. We propose that loss of RhoBTB1 early in breast cancer development promotes loss of normal epithelial polarity through reduced METTL7B expression and Golgi fragmentation. We propose that this then contributes to the switch to an invasive phenotype.

Conclusions

RhoBTB1 is important for maintaining the integrity of the Golgi, through regulation of the expression of METTL7B. In keeping with this, loss of RhoBTB1 expression leads to fragmentation of the Golgi. Restoration of normal RhoBTB1 levels to T47D breast cancer cells restores Golgi integrity and leads to a dramatic decrease in their invasive capacity.

Acknowledgements

We would like to thank the Wolfson Bioimaging Facility for their technical assistance with the cell imaging.

Funding

This study was funded by the Worldwide Cancer Research; Grant number 08–0409.

Availability of data and materials

All of the data supporting our findings can be found in the main paper. All of the plasmids generated in this study are available on request.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

C.M.M. and H.M. designed the study. C.M.M. performed the experimental work. H.M. wrote the manuscript. Both authors have contributed to the data analysis, discussions and interpretations of the results. Both authors have read and approved the final manuscript.
Not applicable.
Not applicable.
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.
Literatur
1.
Zurück zum Zitat Jaffe AB, Hall A. Rho GTPases: biochemistry and biology. Annu Rev Cell Dev Biol. 2005;21:247–69.CrossRefPubMed Jaffe AB, Hall A. Rho GTPases: biochemistry and biology. Annu Rev Cell Dev Biol. 2005;21:247–69.CrossRefPubMed
3.
4.
Zurück zum Zitat Wherlock M, Mellor H. The Rho GTPase family: a racs to wrchs story. J Cell Sci. 2002;115(Pt 2):239–40.PubMed Wherlock M, Mellor H. The Rho GTPase family: a racs to wrchs story. J Cell Sci. 2002;115(Pt 2):239–40.PubMed
5.
Zurück zum Zitat Ramos S, Khademi F, Somesh BP, Rivero F. Genomic organization and expression profile of the small GTPases of the RhoBTB family in human and mouse. Gene. 2002;298(2):147–57.CrossRefPubMed Ramos S, Khademi F, Somesh BP, Rivero F. Genomic organization and expression profile of the small GTPases of the RhoBTB family in human and mouse. Gene. 2002;298(2):147–57.CrossRefPubMed
6.
Zurück zum Zitat Ji W, Rivero F. Atypical Rho GTPases of the RhoBTB Subfamily: Roles in Vesicle Trafficking and Tumorigenesis. Cells. 2016;5(2). Ji W, Rivero F. Atypical Rho GTPases of the RhoBTB Subfamily: Roles in Vesicle Trafficking and Tumorigenesis. Cells. 2016;5(2).
7.
Zurück zum Zitat Hamaguchi M, Meth JL, von Klitzing C, Wei W, Esposito D, Rodgers L, et al. DBC2, a candidate for a tumor suppressor gene involved in breast cancer. Proc Natl Acad Sci U S A. 2002;99(21):13647–52.CrossRefPubMedPubMedCentral Hamaguchi M, Meth JL, von Klitzing C, Wei W, Esposito D, Rodgers L, et al. DBC2, a candidate for a tumor suppressor gene involved in breast cancer. Proc Natl Acad Sci U S A. 2002;99(21):13647–52.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Freeman SN, Cress WD. RhoBTB2 (DBC2) comes of age as a multifunctional tumor suppressor. Cancer Biol Ther. 2010;10(11):1123–5.CrossRefPubMed Freeman SN, Cress WD. RhoBTB2 (DBC2) comes of age as a multifunctional tumor suppressor. Cancer Biol Ther. 2010;10(11):1123–5.CrossRefPubMed
9.
Zurück zum Zitat Knowles MA, Aveyard JS, Taylor CF, Harnden P, Bass S. Mutation analysis of the 8p candidate tumour suppressor genes DBC2 (RHOBTB2) and LZTS1 in bladder cancer. Cancer Lett. 2005;225(1):121–30.CrossRefPubMed Knowles MA, Aveyard JS, Taylor CF, Harnden P, Bass S. Mutation analysis of the 8p candidate tumour suppressor genes DBC2 (RHOBTB2) and LZTS1 in bladder cancer. Cancer Lett. 2005;225(1):121–30.CrossRefPubMed
10.
Zurück zum Zitat Mao H, Qu X, Yang Y, Zuo W, Bi Y, Zhou C, et al. A novel tumor suppressor gene RhoBTB2 (DBC2): frequent loss of expression in sporadic breast cancer. Mol Carcinog. 2010;49(3):283–9.PubMed Mao H, Qu X, Yang Y, Zuo W, Bi Y, Zhou C, et al. A novel tumor suppressor gene RhoBTB2 (DBC2): frequent loss of expression in sporadic breast cancer. Mol Carcinog. 2010;49(3):283–9.PubMed
11.
Zurück zum Zitat Tang W, Wang C, Fu F, Chen Q. RhoBTB2 gene in breast cancer is silenced by promoter methylation. Int J Mol Med. 2014;33(3):722–8.PubMed Tang W, Wang C, Fu F, Chen Q. RhoBTB2 gene in breast cancer is silenced by promoter methylation. Int J Mol Med. 2014;33(3):722–8.PubMed
12.
Zurück zum Zitat Beder LB, Gunduz M, Ouchida M, Gunduz E, Sakai A, Fukushima K, et al. Identification of a candidate tumor suppressor gene RHOBTB1 located at a novel allelic loss region 10q21 in head and neck cancer. J Cancer Res Clin Oncol. 2006;132(1):19–27.CrossRefPubMed Beder LB, Gunduz M, Ouchida M, Gunduz E, Sakai A, Fukushima K, et al. Identification of a candidate tumor suppressor gene RHOBTB1 located at a novel allelic loss region 10q21 in head and neck cancer. J Cancer Res Clin Oncol. 2006;132(1):19–27.CrossRefPubMed
13.
Zurück zum Zitat Xu RS, Wu XD, Zhang SQ, Li CF, Yang L, Li DD, et al. The tumor suppressor gene RhoBTB1 is a novel target of miR-31 in human colon cancer. Int J Oncol. 2013;42(2):676–82.PubMed Xu RS, Wu XD, Zhang SQ, Li CF, Yang L, Li DD, et al. The tumor suppressor gene RhoBTB1 is a novel target of miR-31 in human colon cancer. Int J Oncol. 2013;42(2):676–82.PubMed
14.
Zurück zum Zitat Aspenstrom P, Fransson A, Saras J. Rho GTPases have diverse effects on the organization of the actin filament system. Biochem J. 2004;377(Pt 2):327–37.CrossRefPubMedPubMedCentral Aspenstrom P, Fransson A, Saras J. Rho GTPases have diverse effects on the organization of the actin filament system. Biochem J. 2004;377(Pt 2):327–37.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Perez-Torrado R, Yamada D, Defossez PA. Born to bind: the BTB protein-protein interaction domain. Bioessays. 2006;28(12):1194–202.CrossRefPubMed Perez-Torrado R, Yamada D, Defossez PA. Born to bind: the BTB protein-protein interaction domain. Bioessays. 2006;28(12):1194–202.CrossRefPubMed
16.
Zurück zum Zitat McKinnon CM, Lygoe KA, Skelton L, Mitter R, Mellor H. The atypical Rho GTPase RhoBTB2 is required for expression of the chemokine CXCL14 in normal and cancerous epithelial cells. Oncogene. 2008;27(54):6856–65.CrossRefPubMed McKinnon CM, Lygoe KA, Skelton L, Mitter R, Mellor H. The atypical Rho GTPase RhoBTB2 is required for expression of the chemokine CXCL14 in normal and cancerous epithelial cells. Oncogene. 2008;27(54):6856–65.CrossRefPubMed
17.
Zurück zum Zitat Blaschke RJ, Howlett AR, Desprez PY, Petersen OW, Bissell MJ. Cell differentiation by extracellular matrix components. Methods Enzymol. 1994;245:535–56.CrossRefPubMed Blaschke RJ, Howlett AR, Desprez PY, Petersen OW, Bissell MJ. Cell differentiation by extracellular matrix components. Methods Enzymol. 1994;245:535–56.CrossRefPubMed
18.
Zurück zum Zitat Debnath J, Muthuswamy SK, Brugge JS. Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods. 2003;30(3):256–68.CrossRefPubMed Debnath J, Muthuswamy SK, Brugge JS. Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods. 2003;30(3):256–68.CrossRefPubMed
19.
Zurück zum Zitat Demaison C, Parsley K, Brouns G, Scherr M, Battmer K, Kinnon C, et al. High-level transduction and gene expression in hematopoietic repopulating cells using a human immunodeficiency virus type 1-based lentiviral vector containing an internal spleen focus forming virus promoter. Hum Gene Ther. 2002;13(7):803–13.CrossRefPubMed Demaison C, Parsley K, Brouns G, Scherr M, Battmer K, Kinnon C, et al. High-level transduction and gene expression in hematopoietic repopulating cells using a human immunodeficiency virus type 1-based lentiviral vector containing an internal spleen focus forming virus promoter. Hum Gene Ther. 2002;13(7):803–13.CrossRefPubMed
20.
Zurück zum Zitat Wu CC, MacCoss MJ, Mardones G, Finnigan C, Mogelsvang S, Yates 3rd JR, et al. Organellar proteomics reveals Golgi arginine dimethylation. Mol Biol Cell. 2004;15(6):2907–19.CrossRefPubMedPubMedCentral Wu CC, MacCoss MJ, Mardones G, Finnigan C, Mogelsvang S, Yates 3rd JR, et al. Organellar proteomics reveals Golgi arginine dimethylation. Mol Biol Cell. 2004;15(6):2907–19.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Radeke HS, Digits CA, Casaubon RL, Snapper ML. Interactions of (−)-ilimaquinone with methylation enzymes: implications for vesicular-mediated secretion. Chem Biol. 1999;6(9):639–47.CrossRefPubMed Radeke HS, Digits CA, Casaubon RL, Snapper ML. Interactions of (−)-ilimaquinone with methylation enzymes: implications for vesicular-mediated secretion. Chem Biol. 1999;6(9):639–47.CrossRefPubMed
22.
Zurück zum Zitat Zehmer JK, Bartz R, Liu P, Anderson RG. Identification of a novel N-terminal hydrophobic sequence that targets proteins to lipid droplets. J Cell Sci. 2008;121(Pt 11):1852–60.CrossRefPubMedPubMedCentral Zehmer JK, Bartz R, Liu P, Anderson RG. Identification of a novel N-terminal hydrophobic sequence that targets proteins to lipid droplets. J Cell Sci. 2008;121(Pt 11):1852–60.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Han L, Hou L, Song J, Lin D, Wu L, Ge Y, et al. Decreased expression of the DBC2 gene and its clinicopathological significance in breast cancer: correlation with aberrant DNA methylation. Biotechnol Lett. 2013;35(8):1175–81.CrossRefPubMed Han L, Hou L, Song J, Lin D, Wu L, Ge Y, et al. Decreased expression of the DBC2 gene and its clinicopathological significance in breast cancer: correlation with aberrant DNA methylation. Biotechnol Lett. 2013;35(8):1175–81.CrossRefPubMed
24.
Zurück zum Zitat Ohadi M, Totonchi M, Maguire P, Lindblom A, Habibi R, Alavi BA, et al. Mutation analysis of the DBC2 gene in sporadic and familial breast cancer. Acta Oncol. 2007;46(6):770–2.CrossRefPubMed Ohadi M, Totonchi M, Maguire P, Lindblom A, Habibi R, Alavi BA, et al. Mutation analysis of the DBC2 gene in sporadic and familial breast cancer. Acta Oncol. 2007;46(6):770–2.CrossRefPubMed
25.
Zurück zum Zitat Soule HD, Maloney TM, Wolman SR, Peterson Jr WD, Brenz R, McGrath CM, et al. Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res. 1990;50(18):6075–86.PubMed Soule HD, Maloney TM, Wolman SR, Peterson Jr WD, Brenz R, McGrath CM, et al. Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res. 1990;50(18):6075–86.PubMed
26.
Zurück zum Zitat Briand P, Petersen OW, Van Deurs B. A new diploid nontumorigenic human breast epithelial cell line isolated and propagated in chemically defined medium. In Vitro Cell Dev Biol. 1987;23(3):181–8.CrossRefPubMed Briand P, Petersen OW, Van Deurs B. A new diploid nontumorigenic human breast epithelial cell line isolated and propagated in chemically defined medium. In Vitro Cell Dev Biol. 1987;23(3):181–8.CrossRefPubMed
27.
Zurück zum Zitat Yadav S, Puri S, Linstedt AD. A primary role for Golgi positioning in directed secretion, cell polarity, and wound healing. Mol Biol Cell. 2009;20(6):1728–36.CrossRefPubMedPubMedCentral Yadav S, Puri S, Linstedt AD. A primary role for Golgi positioning in directed secretion, cell polarity, and wound healing. Mol Biol Cell. 2009;20(6):1728–36.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Yadav S, Linstedt AD. Golgi positioning. Cold Spring Harb Perspect Biol. 2011;3(5). Yadav S, Linstedt AD. Golgi positioning. Cold Spring Harb Perspect Biol. 2011;3(5).
30.
Zurück zum Zitat Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol. 2005;15(5):378–86.CrossRefPubMed Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol. 2005;15(5):378–86.CrossRefPubMed
31.
Zurück zum Zitat Rivero F, Dislich H, Glockner G, Noegel AA. The dictyostelium discoideum family of Rho-related proteins. Nucleic Acids Res. 2001;29(5):1068–79.CrossRefPubMedPubMedCentral Rivero F, Dislich H, Glockner G, Noegel AA. The dictyostelium discoideum family of Rho-related proteins. Nucleic Acids Res. 2001;29(5):1068–79.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Genschik P, Sumara I, Lechner E. The emerging family of CULLIN3-RING ubiquitin ligases (CRL3s): cellular functions and disease implications. EMBO J. 2013;32(17):2307–20.CrossRefPubMedPubMedCentral Genschik P, Sumara I, Lechner E. The emerging family of CULLIN3-RING ubiquitin ligases (CRL3s): cellular functions and disease implications. EMBO J. 2013;32(17):2307–20.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Berthold J, Schenkova K, Ramos S, Miura Y, Furukawa M, Aspenstrom P, et al. Characterization of RhoBTB-dependent Cul3 ubiquitin ligase complexes--evidence for an autoregulatory mechanism. Exp Cell Res. 2008;314(19):3453–65.CrossRefPubMedPubMedCentral Berthold J, Schenkova K, Ramos S, Miura Y, Furukawa M, Aspenstrom P, et al. Characterization of RhoBTB-dependent Cul3 ubiquitin ligase complexes--evidence for an autoregulatory mechanism. Exp Cell Res. 2008;314(19):3453–65.CrossRefPubMedPubMedCentral
34.
35.
Zurück zum Zitat Kelly KF, Daniel JM. POZ for effect--POZ-ZF transcription factors in cancer and development. Trends Cell Biol. 2006;16(11):578–87.CrossRefPubMed Kelly KF, Daniel JM. POZ for effect--POZ-ZF transcription factors in cancer and development. Trends Cell Biol. 2006;16(11):578–87.CrossRefPubMed
36.
Zurück zum Zitat Espinosa EJ, Calero M, Sridevi K, Pfeffer SR. RhoBTB3: a Rho GTPase-family ATPase required for endosome to Golgi transport. Cell. 2009;137(5):938–48.CrossRefPubMedPubMedCentral Espinosa EJ, Calero M, Sridevi K, Pfeffer SR. RhoBTB3: a Rho GTPase-family ATPase required for endosome to Golgi transport. Cell. 2009;137(5):938–48.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Lu A, Pfeffer SR. Golgi-associated RhoBTB3 targets cyclin E for ubiquitylation and promotes cell cycle progression. J Cell Biol. 2013;203(2):233–50.CrossRefPubMedPubMedCentral Lu A, Pfeffer SR. Golgi-associated RhoBTB3 targets cyclin E for ubiquitylation and promotes cell cycle progression. J Cell Biol. 2013;203(2):233–50.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Mowbrey K, Dacks JB. Evolution and diversity of the Golgi body. FEBS Lett. 2009;583(23):3738–45.CrossRefPubMed Mowbrey K, Dacks JB. Evolution and diversity of the Golgi body. FEBS Lett. 2009;583(23):3738–45.CrossRefPubMed
40.
Zurück zum Zitat Puthenveedu MA, Bachert C, Puri S, Lanni F, Linstedt AD. GM130 and GRASP65-dependent lateral cisternal fusion allows uniform Golgi-enzyme distribution. Nat Cell Biol. 2006;8(3):238–48.CrossRefPubMed Puthenveedu MA, Bachert C, Puri S, Lanni F, Linstedt AD. GM130 and GRASP65-dependent lateral cisternal fusion allows uniform Golgi-enzyme distribution. Nat Cell Biol. 2006;8(3):238–48.CrossRefPubMed
41.
Zurück zum Zitat Diao A, Rahman D, Pappin DJ, Lucocq J, Lowe M. The coiled-coil membrane protein golgin-84 is a novel rab effector required for Golgi ribbon formation. J Cell Biol. 2003;160(2):201–12.CrossRefPubMedPubMedCentral Diao A, Rahman D, Pappin DJ, Lucocq J, Lowe M. The coiled-coil membrane protein golgin-84 is a novel rab effector required for Golgi ribbon formation. J Cell Biol. 2003;160(2):201–12.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Kupfer A, Louvard D, Singer SJ. Polarization of the Golgi apparatus and the microtubule-organizing center in cultured fibroblasts at the edge of an experimental wound. Proc Natl Acad Sci U S A. 1982;79:2603–07. Kupfer A, Louvard D, Singer SJ. Polarization of the Golgi apparatus and the microtubule-organizing center in cultured fibroblasts at the edge of an experimental wound. Proc Natl Acad Sci U S A. 1982;79:2603–07.
43.
Zurück zum Zitat Baschieri F, Confalonieri S, Bertalot G, Di Fiore PP, Dietmaier W, Leist M, et al. Spatial control of Cdc42 signalling by a GM130-RasGRF complex regulates polarity and tumorigenesis. Nat Commun. 2014;5:4839.CrossRefPubMedPubMedCentral Baschieri F, Confalonieri S, Bertalot G, Di Fiore PP, Dietmaier W, Leist M, et al. Spatial control of Cdc42 signalling by a GM130-RasGRF complex regulates polarity and tumorigenesis. Nat Commun. 2014;5:4839.CrossRefPubMedPubMedCentral
Metadaten
Titel
The tumor suppressor RhoBTB1 controls Golgi integrity and breast cancer cell invasion through METTL7B
verfasst von
Caroline M. McKinnon
Harry Mellor
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2017
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3138-3

Weitere Artikel der Ausgabe 1/2017

BMC Cancer 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.