SlideShare a Scribd company logo
1 of 19
Download to read offline
DIAPH3 Governs the Cellular Transition to the Amoeboid Tumour 
Phenotype 
The Harvard community has made this article openly available. 
Please share how this access benefits you. Your story matters. 
Citation Hager, Martin H., Samantha Morley, Diane R. Bielenberg, 
Sizhen Gao, Matteo Morello, Ilona N. Holcomb, Wennuan Liu, 
et al. 2012. Diaph3 governs the cellular transition to the 
amoeboid tumour phenotype. EMBO Molecular Medicine 4(8): 
743-760. 
Published Version doi:10.1002/emmm.201200242 
Accessed October 8, 2013 10:44:17 PM EDT 
Citable Link http://nrs.harvard.edu/urn-3:HUL.InstRepos:10589807 
Terms of Use This article was downloaded from Harvard University's DASH 
repository, and is made available under the terms and conditions 
applicable to Other Posted Material, as set forth at 
http://nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- 
use#LAA 
(Article begins on next page)
DIAPH3 governs the cellular transition 
to the amoeboid tumour phenotype 
Martin H. Hager1,2y,z, Samantha Morley1,2z, Diane R. Bielenberg2,3, Sizhen Gao4, Matteo Morello1,2,5, 
Ilona N. Holcomb6, Wennuan Liu7, Ghassan Mouneimne4, Francesca Demichelis8,9, Jayoung Kim1,2,5, 
Keith R. Solomon1,10, Rosalyn M. Adam1,2, William B. Isaacs11, Henry N. Higgs12, Robert L. Vessella13, 
Dolores Di Vizio1,2,5, Michael R. Freeman1,2,5,14* 
Keywords: cytoskeleton; EGFR; 
endocytosis; mesenchymal-to-amoeboid 
transition; metastasis 
DOI 10.1002/emmm.201200242 
Received November 23, 2011 
Revised March 19, 2012 
Accepted March 28, 2012 
Therapies for most malignancies are generally ineffective once metastasis occurs. 
While tumour cells migrate through tissues using diverse strategies, the signalling 
networks controlling such behaviours in human tumours are poorly understood. 
Here we define a role for the Diaphanous-related formin-3 (DIAPH3) as a non-canonical 
regulator of metastasis that restrains conversion to amoeboid cell 
behaviour in multiple cancer types. The DIAPH3 locus is close to RB1, within a 
narrow consensus region of deletion on chromosome 13q in prostate, breast and 
hepatocellular carcinomas. DIAPH3 silencing in human carcinoma cells destabi-lized 
microtubules and induced defective endocytic trafficking, endosomal 
accumulation of EGFR, and hyperactivation of EGFR/MEK/ERK signalling. Silencing 
also evoked amoeboid properties, increased invasion and promoted metastasis in 
mice. In human tumours, DIAPH3 down-regulation was associated with aggressive 
or metastatic disease. DIAPH3-silenced cells were sensitive to MEK inhibition, but 
showed reduced sensitivity to EGFR inhibition. These findings have implications for 
understanding mechanisms of metastasis, and suggest that identifying patients 
with chromosomal deletions at DIAPH3 may have prognostic value. 
INTRODUCTION 
Improving clinical options for metastatic disease requires an 
understanding of the molecular basis for the complex 
behaviours of metastatic cells. To date, however, few genetic 
lesions in human tumours that promote selection of metastatic 
variants have been identified. 
Tumour cell migration can lead to metastatic dissemination. 
The ability of carcinoma cells to transition from an adherent, 
epithelial phenotype to a migratory, mesenchymal phenotype 
(the epithelial-to-mesenchymal transition, EMT) is well estab-lished, 
as are many signalling networks responsible for this 
transformation (Kalluri & Weinberg, 2009). However, it has 
recently been recognized that cancer cells can further transition 
Research Article 
DIAPH3 and the amoeboid phenotype 
(1) Urological Diseases Research Center, Children’s Hospital Boston, Boston, 
MA, USA 
(2) Department of Surgery, Harvard Medical School, Boston, MA, USA 
(3) Vascular Biology Program, Children’s Hospital Boston, Boston, MA, USA 
(4) Department of Cell Biology, Harvard Medical School, Boston, MA, USA 
(5) Division of Cancer Biology and Therapeutics, Samuel Oschin Compre-hensive 
Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 
USA 
(6) Department of Pathology, Stanford University School of Medicine, 
Stanford, CA, USA 
(7) Center for Cancer Genomics, Wake Forest University, Winston-Salem, NC, 
USA 
(8) Department of Pathology and Laboratory Medicine, Weill Cornell Medical 
College, New York, NY, USA 
(9) Centre for Integrative Biology, University of Trento, Trento, Italy 
(10) Department of Orthopaedic Surgery, Children’s Hospital Boston, 
Harvard Medical School, Boston, MA, USA 
(11) Department of Urology and Oncology, Johns Hopkins School of 
Medicine, Baltimore, MD, USA 
(12) Department of Biochemistry, Dartmouth Medical School, Hanover, NH, USA 
(13) Department of Urology, University of Washington, Seattle, WA, USA 
(14) Department of Biological Chemistry and Molecular Pharmacology, 
Harvard Medical School, Boston, MA, USA 
*Corresponding author: Tel: þ1 310 423 7069; Fax: þ1 310 423 0139; 
E-mail: michael.freeman@cshs.org 
y Present address: R&D Division, Oncology Research Laboratories, Daiichi 
Sankyo Europe, Munich, Germany 
zThese authors contributed equally to this work. 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 743
to an ‘amoeboid’ phenotype (called the mesenchymal-to-amoeboid 
transition, MAT), characterized by a rounded 
morphology, extensive actomyosin contractility, rapid exten-sion 
and retraction of membrane protrusions (blebs), and 
reduced dependence on proteolysis for migration (Friedl  Wolf, 
2010). Amoeboid cells are behaviourally plastic, move rapidly 
through extracellular matrices by deforming their shape, and 
express surface proteins more symmetrically than mesenchymal 
cells, thus facilitating opportunistic responses to changes in 
the microenvironment (Schmidt  Friedl, 2010). While such 
characteristics enable cells to readily migrate through extra-cellular 
matrix (ECM), the clinical impact of the amoeboid 
phenotype is unknown. Additionally, EMT (Kalluri  Weinberg, 
2009) and MAT (Wolf et al, 2003) are reversible processes, and 
the ability of tumour cells to switch between these varied states 
presents challenges in defining critical signalling nodes where 
metastasis might be inhibited. While the amoeboid phenotype is 
potentially relevant to aggressive cancer (Sanz-Moreno  
Marshall, 2010), it has not been well studied in human tumours. 
Chromosome 13 is frequently altered in cancer, and the q-arm 
suffers predominantly from deletions (Beroukhim et al, 2010). 
The RB1 locus at 13q14.2 is thought to be the dominant tumour 
suppressor affected by copy number alterations. However, 
reports speculate that another tumour suppressor resides in 
this region, whose loss is associated with breast (Kainu et al, 
2000) and invasive prostate (Dong et al, 2000) cancers, 
and whose restoration can inhibit metastatic dissemination 
(Hosoki et al, 2002). 
Proximal to RB1, at 13q21.2, is the DIAPH3 locus encoding the 
protein Diaphanous-related formin-3 [DIAPH3, (Peng et al, 
2003)]. This protein belongs to a family of formin orthologs 
(Chalkia et al, 2008) that nucleate, elongate and bundle linear 
actin filaments and regulate microtubule dynamics (Bartolini  
Gundersen, 2010; Gaillard et al, 2011; Goode  Eck, 2007). 
Formins are required for such fundamental processes as 
endocytic trafficking, mitosis, cell polarity and migration (Goode 
 Eck, 2007). Despite the deregulation of these events in 
pathological states such as cancer, the role of DIAPH3 in disease 
is unexplored.We recently reported that genomic loss at DIAPH3 
was linked to metastatic prostate cancer (Di Vizio et al, 2009). 
However, given the propensity for chromosomal loss along 13q in 
multiple cancers (Beroukhim et al, 2010), whether DIAPH3 
deletions are driver or passenger lesions remained unresolved. 
We now demonstrate that genomic loss at DIAPH3 is 
associated with multiple tumour types, and that DIAPH3 resides 
at an important signalling node that controls the amoeboid 
phenotype. We show that DIAPH3 silencing disrupts micro-tubules, 
impairs endocytosis and prevents EGFR downregulation, 
thereby enhancing EGFR activity and that of its downstream 
effectors MEK and ERK. DIAPH3 deficiency also promotes 
motility, invasion and experimental metastasis and significantly 
correlates with aggressive disease in human tumours. 
RESULTS 
Loss of DIAPH3 with tumour progression 
To test whether genomic loss at DIAPH3 is a driver or passenger 
effect consequent to the general instability of chromosome 13q, 
we employed the genomic identification of significant targets in 
cancer (GISTIC) platform (Beroukhim et al, 2010). GISTIC 
identified regions of somatic copy number alterations (SCNA) in 
a pooled analysis of 26 different cancer types, most of which are 
not associated with previously validated tumour suppressor 
genes. We hypothesized that significant (q-value 0.25) focal 
SCNA harbouring a tumour suppressor would emerge as a 
consensus area across these cancers. Among the deletion peaks 
on chromosome 13, one containing three genes, protocadherin 
17 (PCDH17), tudor domain-containing protein 3 (TDRD3) and 
DIAPH3, was located in a region common to three carcinomas 
(prostate, breast and hepatocellular, Fig 1A). DIAPH3, near the 
centre of this consensus region, was deleted in 32% of prostate, 
46% of breast and 31% of hepatocellular carcinomas (Support-ing 
Information Fig S1A). Together with the associated q-value 
range of 0.052q0.132, these data suggest that DIAPH3 
deletions are enriched by selective pressure. In support of this 
hypothesis, Oncomine expression profiles indicated DIAPH3 
mRNA levels were lower in high stage prostate cancer (PCa) and 
in hepatocellular and invasive breast carcinomas than in normal 
tissue [Fig 1B–D; (Finak et al, 2008; Li et al, 2006; Mas et al, 
2009)]. 
Given the prevalence of DIAPH3 lesions in multiple tumour 
types, we examined DIAPH3 status in metastases, using PCa as a 
representative disease model. Metastatic PCa lesions can 
originate from a single cell (Liu et al, 2009) and may lie 
dormant for many years (Wikman et al, 2008). For these 
reasons, we characterized the DIAPH3 status of disseminated 
tumour cells (DTCs) isolated from bone marrow of PCa patients 
(Holcomb et al, 2008). Deletions were more frequent in DTCs 
with either prostate-confined (31%) or metastatic (46%) disease 
Research Article 
DIAPH3 and the amoeboid phenotype 
Figure 1. Genomic loss at DIAPH3 in invasive and metastatic tumours. 
A. The DIAPH3 locus on chromosome 13q21.2 is located in a focal peak region of significant deletion (q0.25) common in prostate, hepatocellular and breast 
carcinomas. Only three genes, PCDH17, TDRD3 and DIAPH3, are focally deleted in all cancer subtypes. 
B-D. DIAPH3 mRNA levels in prostate (PCa, stage T2/T3), hepatocellular and invasive breast carcinomas compared to normal tissue. 
E. Frequency of loss at DIAPH3 (grey line) in DTCs aspirated from bone marrow of patients with prostate-confined cancer (n¼48, blue) and advanced PCa 
(n¼11, red). Determined by array CGH and compared to copy number status in the primary tumours (n¼9, green). The chromosome band for each genomic 
position is shown; tick marks above indicate gene locations on chromosome 13. 
F. DIAPH3 copy number status grouped by Gleason score and compared with metastases. 
G. DIAPH3 copy number analysis in matched primary tumours and metastases (Met) reveals significant loss in metastatic disease. 
H. DIAPH3 gene copy number in PCa samples grouped by Gleason Score in comparison to metastases (Met). 
 
744  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
Research Article 
Martin H. Hager et al. 
B C Hepatocellular carcinoma D Breast carcinoma 
n=6 n=53 
N 
Normalized expression 
units DIAPH3 
0.5 
-0.5 
-1.5 
invasive 
BCa 
Normalized expression 
units DIAPH3 
-1.8 
-2.0 
-2.2 
-2.4 
n=19 n=38 
N HCa 
Prostate carcinoma 
Normalized expression 
-1.2 
units DIAPH3 
n=4 n=4 n=12 
T2 
-1.5 
-1.8 
-2.1 
N T3 
PCa 
A 
n=53 n=102 n=12 n=12 n=36 
Advanced 
Disease 
Prostate-confined 
Disease 
-0.5 Met 
Gleason Score 
0.2 
0.1 
0 
-0.1 
-0.2 
-0.3 
-0.4 
DIAPH3 log2 copy 
number units 
-0.5 
6 7 8 9 
0.1 
20 
10 
F G 
DIAPH3 log2 copy 
number units 
0 
-0.1 
-0.2 
-0.3 
-0.4 
n=181 n=37 
Primary 
Met 
Site 
80 
70 
60 
50 
40 
30 
20 
10 
% of patients with deletion 
at DIAPH3 locus 
Grouped by 
Gleason Score 
n = 24 
n = 63 
n = 43 
6 7 8 
Gleason Score 
Metastasis 
0 
n = 18 
Mbp H 
50 
40 
30 
RB1 DIAPH3 
0 
50 55 60 65 70 75 
% of samples 
Primary 
Tumour 
Disseminated 
Tumour Cells 
E 
Figure 1. 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 745
than in primary tumours (10%, Fig 1E), suggesting that DIAPH3 
loss is related to dissemination from the primary site. 
To examine this possibility, we evaluated DIAPH3 copy 
number in primary tumours and metastases of 148 prostate 
cancers by Affymetrix Genome-Wide Human SNP Array 6.0 (Liu 
et al, 2009). Loss at DIAPH3 was increasingly prevalent with 
progression, occurring in 25% of low grade (Gleason score 6), 
33% of intermediate grade (Gleason score 7), 40% of high grade 
(Gleason score 8), and 61% of metastatic tumours (Fig 1F, 
Supporting Information Fig S1B). Oncomine copy number data 
confirmed DIAPH3 genomic loss and its significant correlation 
with disease progression, which increased from primary tumour 
to metastatic lesions and with increasing Gleason score [Fig 1G 
and H; (Taylor et al, 2010)]. 
DIAPH3 knockdown induces an amoeboid phenotype in 
transformed cells 
In order to examine the biological consequences of DIAPH3 loss, 
we employed RNAi against DIAPH3 with multiple independent 
shRNAs or siRNAs (Supporting Information Fig S2A, Fig 6C), 
which efficiently reduced DIAPH3 protein levels but did not 
affect the related proteins DIAPH1 and DIAPH2 (Supporting 
Information Fig S2B). Because DIAPH3 loss was observed in 
breast cancers (Fig 1A and D), we employed the disease-relevant 
human mammary epithelial cell (HMEC) model. While stable 
DIAPH3 knockdown did not alter HMEC cell morphology 
in 2D culture, it modestly enhanced proliferation (Fig 2A, 
Supporting Information Fig S2C and D). In reconstituted 
basement membrane gels (3D culture), DIAPH3 silencing 
induced enlarged acinar structures with aberrant architecture 
and distribution of Laminin V (Fig 2A, Supporting Information 
Fig S3A and B), resembling that seen with ErbB2 activation 
(Debnath et al, 2002). These results suggest that in normal 
epithelia, DIAPH3 may act to suppress oncogenic behaviour. 
Next, we investigated the consequences of DIAPH3 loss in the 
context of HRAS-induced cellular transformation. We generated 
stable populations of HRASV12-transformed HMECs in which 
DIAPH3 was silenced (Supporting Information Fig S3C). In 2D 
culture, HMEC-HRASV12 cells appeared spindle-shaped, con-sistent 
with EMT (Fig 2B, top). In contrast, DIAPH3-depleted 
HMEC-HRASV12 cells were refractile, with rounded morpholo-gies 
(Fig 2B). Time-lapse video microscopy demonstrated that 
these cells displayed abundant, reversible eruption of blebs, 
reduced cell–cell contact, and increased rates of random 
motility, in comparison to HMEC-HRASV12 cells (Fig 2C 
and D, Supporting Information Movies S1, S2). These features 
are indicative of amoeboid behaviour (Wolf et al, 2003). 
In 3D culture, HMEC-HRASV12 produced large cell aggregates 
(acini) with penetration of the matrix by cords of spindle-shaped 
cells (Fig 2B, top inset, Supporting Information Fig S3D). In 
contrast, DIAPH3-deficient HRASV12 cells produced acini that 
progressively and dramatically lost structural integrity, coin-ciding 
with extensive migration of round cells into the matrix 
(Fig 2B, lower insets, Supporting Information Fig S3D). 
Significantly, while HRASV12-HMEC cells were poorly invasive 
through collagen I, DIAPH3 knockdown in this cell background 
promoted invasion (Fig 2E). These findings indicate that, when 
combined with activated HRAS, DIAPH3 down-regulation 
promotes an amoeboid phenotype and increases invasion. 
To determine whether this amoeboid transition can be 
similarly induced in genuine cancer cells, we silenced DIAPH3 
in DU145 PCa cells. As in the genetically defined HRASV12- 
HMEC model, DIAPH3 knockdown in DU145 cells induced a 
rounded morphology and formation of membrane blebs when 
cells were embedded in collagen (Fig 2F). In line with this 
promotion of amoeboid behaviour (Fig 2G), DIAPH3 knock-down 
enhanced the invasiveness of DU145 cells (Fig 2H, 
Supporting Information Fig S3E). DIAPH3-deficient cells were 
hypersensitive to chemo-attractants, including EGF. 
Amoeboid cells display weakened substrate adhesions, 
allowing high cell velocities (Schmidt  Friedl, 2010). This fast 
‘gliding’ mode can be driven by Rho-kinase (ROCK)-dependent 
force generation through myosin light chain (MLC2) phosphor-ylation, 
and is distinct from integrin-mediated traction force 
generation characteristic of mesenchymal motility (Friedl  
Wolf, 2010). DIAPH3 silencing promoted focal adhesion 
disassembly, as evidenced by an altered pattern of focal 
adhesion kinase (FAK) phosphorylation [Fig 3A, 3B (Hamadi 
et al, 2005)], and also evoked high levels of phosphorylated 
MYPT1, a ROCK substrate (Fig 3C and D), and MLC2 (Fig 3E). In 
DIAPH3-deficient cells, both activated and total MLC2 were 
enriched at the cell cortex (Fig 3F, Supporting Information 
Fig S4A–C), where cortical MLC2 colocalized with phalloidin. In 
unsilenced cells, cortical MLC2 was less pronounced and 
co-localized insteadwith phalloidin in stress fibres. These findings 
are consistent with the ROCK-mediated cortical localization of 
MLC2 reported to be displayed by amoeboid cells (Pinner  Sahai, 
2008; Sahai  Marshall, 2003; Wyckoff et al, 2006). 
Research Article 
DIAPH3 and the amoeboid phenotype 
Figure 2. DIAPH3 knockdown induces an amoeboid phenotype in transformed cells. 
A. Morphology of DIAPH3-depleted HMEC, compared to control cells. Plastic (2D), basement membrane cultures (3D). Scale¼200 mm. 
B. HRASV12 cells grown in 2D adopt a round, refractile appearance when DIAPH3 is silenced. Scale¼100 mm. HMEC-HRASV12 cells form cell aggregates with cord-like 
protrusions in 3D. DIAPH3 depletion causes junctional instability, with migration of single cells into the surrounding matrix (inset, right). Scale¼200 mm. 
C. Representative trajectories of control (green) and DIAPH3-deficient (red) HMEC-HRASV12 cells. 
D. Quantification of migration speed, determined from C. 
E. DIAPH3-silenced HRASV12-HMEC display increased invasiveness. 
F. Growth of DIAPH3-deficient and control DU145 cells on a thick layer of collagen I. Arrowheads mark cell surface blebbing (top). Visualization of the 
cytoskeleton with phalloidin by immunofluorescence (IF) shows prominent cortical actin in DIAPH3-deficient cells (arrowhead, bottom). Scale¼50 mm. 
G. Increase in amoeboid morphology following expression of DIAPH3 siRNA. 
H. Invasion of collagen I by DU145 cells, with FBS or EGF as chemo-attractants, is increased in DIAPH3-deficient cells (N2 independent trials). See also 
Supporting Information Movies S1 and S2. 
 
746  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
Research Article 
Martin H. Hager et al. 
B 
HMEC HMEC-RasV12 
2D 3D 2D 3D 3D (inset) 
C D Migration E 
Distance (μm) 
Distance (μm) 
Control shRNA 
DIAPH3 shRNA 
Fluorescence 
(RFU X 103) 
Invasion 
50 
40 
30 
20 
10 
0 
Control shRNA 
HMEC 
HRASV12 
Control shRNA 
DIAPH3 shRNA 
4 
3 
2 
1 
0 
Speed 
(μm/min) 
HMEC 
HRASV12 
DU145 Invasion 
G Morphology 
F H 
Control shRNA DIAPH3 shRNA 
Control shRNA 
DIAPH3 shRNA 
FBS 6h FBS 12h EGF 12h 
Fluorescence 
(RFU X 103) 
80 
60 
40 
20 
0 
DIAPH3 shRNA 
A 
F-actin F-actin 
Control shRNA 
DIAPH3 shRNA1 
DIAPH3 shRNA2 
Control shRNA 
DIAPH3 shRNA1 
DIAPH3 shRNA2 
Control shRNA 
DIAPH3 shRNA1 
DIAPH3 shRNA2 
Control shRNA 
DIAPH3 shRNA1 
DIAPH3 shRNA2 
Control shRNA 
DIAPH3 shRNA1 
DIAPH3 shRNA2 
100 
80 
60 
40 
20 
0 
amoeboid cells/field 
(percent of total) 
Control siRNA 
DIAPH3 siRNA 
DU145 
DU145 
Figure 2. 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 747
A B 
Control 
shRNA 
MYPT1 
DIAPH3 regulates EGFR trafficking and microtubule stability 
We next sought to examine the mechanisms underlying 
induction of the amoeboid transition by DIAPH3 knockdown. 
Formins localize to endosomes and regulate vesicular trafficking 
(Fernandez-Borja et al, 2005; Gasman et al, 2003; Wallar et al, 
2007). Because endocytosis governs the spatiotemporal regula-tion 
of receptor tyrosine kinases (RTKs), and chemotactic 
sensitivity to EGF was enhanced in DIAPH3-silenced cells 
(Fig 2H), we hypothesized that DIAPH3 may regulate turnover 
and trafficking of EGFR. Enforced expression of DIAPH3 
markedly accelerated the kinetics of EGFR turnover and receptor 
Control siRNA 
Control shRNA 
DIAPH3 shRNA 
12 
10 
8 
6 
4 
2 
20 
16 
12 
8 
4 
pMLC2 (S19) Phalloidin 
inactivation in response to EGF and under steady-state 
conditions (Fig 4A, 4B). Further supporting a functional 
relationship, DIAPH3 and EGFR co-localized in endosomes 
(unpublished observations). Thus, we examined the influence 
of DIAPH3 on endocytic trafficking of EGFR. In the absence of 
EGF and the presence of endogenous DIAPH3, EGFR co-localized 
at the plasma membrane with the recycling marker 
Rab11 (Fig 4C). In response to ligand, EGFR translocated to a 
peri-nuclear region and co-localized with the early endosome 
marker Rab5 (Fig 4D). In contrast, DIAPH3 silencing promoted 
association of EGFR with endosomes enriched in Rab11, Rab5 
Research Article 
DIAPH3 and the amoeboid phenotype 
Focal adhesions per cell 
DIAPH3 siRNA 
pMLC2 
MLC2 
DIAPH3 
-Actin 
Control 
shRNA 
DIAPH3 
shRNA 
F DU145-Control shRNA DU145-DIAPH3 shRNA 
pMLC2 (S19) Phalloidin 
Dapi Merge Dapi Merge 
C 
ROCK 
DIAPH3 
siRNA 
Control 
siRNA 
pMYPT1 
MYPT1 
pMYPT1 
pMYPT1/MYPT1 
(fold of control) 
0 
ROCK 
DIAPH3 
shRNA 
D 
0 
p0.0001 
Control siRNA 
pFAK (Y397) 
DIAPH3 siRNA 
pFAK (Y397) 
E 
Figure 3. DIAPH3 knockdown promotes biochemical features of the amoeboid transition. 
A. The distribution of focal adhesions in COS7 cells, detected by IF with a phospho-FAK(Y397) antibody, is reduced in DIAPH3-deficient cells. Scale¼20 mm. 
B. Quantitation of focal adhesions in A. 
C-E. Enhanced phosphorylation of MYPT1 (C,D) and MLC2 (E) in DIAPH3-deficient DU145 cells indicates high ROCK activity. 
F. Distribution of active (phosphorylated) MLC2 and F-actin (phalloidin) in DU145 stable lines. Scale¼10mm (N2 independent trials). 
748  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
and EEA1 (Fig 4C–E), even in absence of ligand. These findings 
suggest that loss of DIAPH3 disrupts vesicular transport, leading 
to EGFR accumulation in endosomes. 
To investigate how DIAPH3 affects endosomal trafficking 
of EGFR, we assessed its influence on the actin and micro-tubule 
(MT) cytoskeletons, which govern short- and long-range 
vesicular transport, respectively (Soldati  Schliwa, 2006). While 
rearrangement of the actin cytoskeleton was associated with 
DIAPH3 silencing (Fig 3F, Supporting Information Fig S4C), no 
conspicuous actin defects were detected. Combined with the 
observation that endosomes did not accumulate at the cell cortex 
but were dispersed throughout the cytoplasm in DIAPH3- 
deficient cells (Fig 4C–E), these data suggest that DIAPH3 loss 
does not significantly influence short-range EGFR trafficking. 
In addition to regulating actin dynamics, formins also 
promote MT stability (Bartolini et al, 2008). Enforced DIAPH3 
markedly increased tubulin acetylation (Fig 5A, Supporting 
Information Fig S5A), an indication of MT stabilization (Verhey 
 Gaertig, 2007). Conversely, DIAPH3 silencing decreased 
tubulin acetylation and was associated with MT fragmentation 
(Fig 5B–G, Supporting Information Fig S5B). Notably, treatment 
of cells with the MT depolymerizing agent nocodazole 
phenocopied the accumulation of EGFR in internal vesicles 
and some amoeboid features (Supporting Information Fig S5C 
and D) induced by DIAPH3 knockdown. 
These findings imply that DIAPH3 deficiency disrupts MT 
stability and endosome processing, thereby sequestering and 
preserving EGFR in early endosomes. Disrupted trafficking 
coincided with EGFR hyper-activation, as evidenced by 
increased auto-phosphorylation of activating residues and 
decreased phosphorylation of the inhibitory Thr669 site 
(Fig 6A). EGFR activation was enhanced in DIAPH3-silenced 
cells in response to EGF (Supporting Information Fig S6A). 
DIAPH3 deficiency also increased phosphorylation of the EGFR 
effectors MEK and ERK (Fig 6A and B, Supporting Information 
Fig S6A), which inversely and dose-dependently correlated with 
DIAPH3 levels (Fig 6C, Supporting Information Fig S6B). Even 
under serum-free conditions, DIAPH3 depletion increased ERK 
phosphorylation sixfold (Fig 6D), and potentiated the response 
to EGF (Fig 6E, Supporting Information Fig S6C), with a twofold 
to fourfold increase in phosphorylated ERK at each time point 
post-stimulation. This ligand-independent hyperactivation of 
EGFR and MEK/ERK is consistent with reports linking early 
endosome-localized EGFR to sustained activation of the RAF/ 
MEK/ERK pathway (Sorkin  von Zastrow, 2009). Collectively, 
Research Article 
Martin H. Hager et al. 
these findings support an important role for DIAPH3 in 
regulation of EGFR trafficking and signalling. 
Sensitivity of DIAPH3-deficient cells to anti-neoplastic agents 
Because DIAPH3 silencing induces amoeboid behaviour and 
strongly activates ERK, we tested the relevance of MEK/ERK 
signalling in this scenario. Treatment of DIAPH3-deficient 
DU145, HMEC-HRASV12 and A375P melanoma cells with the 
MEK1/2 inhibitor PD98059 converted cells to a more mesench-ymal 
morphology (Fig 7A, Supporting Information Fig S7A). 
Pharmacologic reversion of amoeboid features could be 
reversed by drug washout (unpublished observations). A375P 
has been used as a model to study amoeboid properties (Sanz- 
Moreno et al, 2008). Collectively, these data indicate that MEK/ 
ERK pathway activation can contribute to amoeboid behaviour 
in diverse cell backgrounds. 
EGFR is a frequent target for therapeutic intervention. 
However, responsiveness to tyrosine kinase inhibitors (TKI, e.g. 
gefitinib)may be dictated by the receptor’s subcellular localization 
(Huang et al, 2009). Because DIAPH3 knockdown modulates 
EGFR localization and activity, we asked whether DIAPH3 
deficiency affects TKI sensitivity. In control DU145 cells, EGF 
stimulation induced amoeboid features (blebbing), which was 
suppressed by gefitinib (Fig 7B and C, Supporting Information 
Movie S3). In contrast, the amoeboid phenotype was constitutive 
and ligand-independent in DIAPH3-silenced cells. Notably, these 
cells were unresponsive to the TKI. These findings suggest that 
DIAPH3 loss alters sensitivity to EGFR inhibitors. 
Taxanes such as paclitaxel and docetaxel are first-line 
chemotherapies for metastatic breast and prostate cancers, 
which we show are prone to DIAPH3 loss (Fig 1). However, 
development of resistance limits their efficacy in patients. 
Taxanes stabilize MTs, suggesting their potential to down-regulate 
EGFR similarly to DIAPH3. We observed, however, that 
EGFR activation persisted in the presence of docetaxel 
(Supporting Information Fig S7B) and paclitaxel (unpublished 
observations), in line with reports that these agents disrupt 
endosomal EGFR trafficking (Sonee et al, 1998). 
DIAPH3 suppresses the amoeboid phenotype 
Induction of the amoeboid phenotype by DIAPH3 knockdown 
suggests that enforced DIAPH3 expression may counteract this 
transition. We stably expressed GFP-DIAPH3 in PC3 PCa cells 
and in U87 glioblastoma cells. Of note, U87 expressed the lowest 
levels of DIAPH3 among the cell lines we tested. U87 and PC3 
Figure 4. DIAPH3 regulates endocytic trafficking of EGFR. 
A. Enforced DIAPH3 accelerates EGFR turnover. COS7 cells expressing empty vector (Vo) or DIAPH3 were serum-depleted overnight, treated with EGF for the 
indicated times, and lysates blotted with the antibodies shown. 
B. GFP-DIAPH3-positive COS7 cells show strongly reduced EGFR levels (lower panel, arrowhead) in comparison with GFP-transfected control cells (upper panel) or 
adjacent untransfected cells. Scale¼20 mm. 
C. Left, Co-localization of Cy3-labeled EGFR with FITC-labelled Rab11 in DU145 cells expressing DIAPH3 siRNAs. Right, Cell peripheries are shown in grey, and 
areas of co-localization in black. Insets show predominance of EGFR at the plasma membrane (top) or in endosomes (bottom). Scale¼20mm. 
D. Left, Co-localization of Cy3-labeled EGFR with FITC-labelled Rab5 upon ligand binding (þEGF). DIAPH3 loss evokes an increase in EGFR internalization in the 
absence of ligand (EGF), as shown by accumulation in endosomes (insets and arrowheads) and co-localization with Rab5. Scale¼20 mm. Right, Quantitation 
of EGFR-enriched endosomes. 
E. Cy3-labeled EGFR, which is localized at the plasma membrane in DU145 cells expressing control siRNA, is internalized and co-localizes with the FITC-labelled 
early endosome marker EEA1 in DU145 cells expressing DIAPH3 siRNAs. Scale¼10mm (N2 independent trials). 
 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 749
Research Article 
DIAPH3 and the amoeboid phenotype 
EGF: 
pEGFR (Y1086) 
EGFR 
DIAPH3 
-actin 
Vo DIAPH3 
A 
B 
C E 
Control siRNA 
DIAPH3 siRNA 
180 
150 
120 
90 
60 
30 
0 
Control siRNA 
- EGF + EGF 
EGFR-positive 
endosomes per cell 
DIAPH3 siRNA 
EGFR Rab11 
Control siRNA 
EGFR EGFR 
Rab5 Rab5 
Merge Merge 
DIAPH3 siRNA 
EGFR 
Rab5 
Merge 
- EGF + EGF 
- EGF + EGF 
EGFR EEA1 
EGFR EEA1 
Common pixels 
Control shRNA 
DIAPH3 shRNA 
EGFR Rab11 
EGFR/GFP-DIAPH3 
GFP-DIAPH3 GFP 
EGFR EGFR/GFP 
EGFR 
D 
EGFR 
Rab5 
Merge 
Figure 4. 
750  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
Research Article 
Martin H. Hager et al. 
Control 
shRNA 
Ac-tubulin 
DIAPH3 
shRNA 
DIAPH3 
-actin 
A 
Control shRNA 
DIAPH3 shRNA 
p = 0.0011 
Ac-tubulin / -actin 
(fold of control) 
1.0 
0.5 
0 
Ac-tubulin 
DIAPH3 
-actin 
Vo 
DIAPH3 
p = 0.038 
-tubulin 
Ac-tubulin / -actin 
(fold of control) 
Vo DIAPH3 
8 
6 
4 
2 
0 
D 
Ac-tubulin 
fluorescence 
Control shRNA 
Tubulin pixels 
(rendered) 
DIAPH3 shRNA 
x 
y 
x 
y 
Pixel intensity, y plane 
Control shRNA 
Distance (pixels), x plane 
12 
9 
6 
0 
100 200 300 400 500 
3 
DIAPH3 shRNA 
C 
B 
F Control shRNA DIAPH3 shRNA G 
Phalloidin Ac-tub Phalloidin Ac-tub 
Control shRNA 
DIAPH3 shRNA 
p = 0.0033 
n = 488 
Ac-tubulin Intensity 
(fold of control) 
1.0 
0.5 
0 
n = 577 
Control shRNA DIAPH3 shRNA 
CTxB Ac-tub CTxB Ac-tub 
E 
Figure 5. DIAPH3 regulates microtubule topology and stability. 
A. Increased acetylated tubulin (Ac-tubulin) in PC3 cells stably expressing DIAPH3. 
B. Reduced Ac-tubulin levels in DU145 cells silenced for DIAPH3. 
C. Left, Ac-tubulin IF showing MT fragmentation in DIAPH3-deficient DU145 cells grown in 3D. Right, MT topology was rendered by ImageJ. Insets, cells stained 
with cholera toxin B (CTxB, green). Scale¼10 mm. 
D. Pixel intensity of C was quantified as a 2D contour plot, as a function of intensity in the x versus y planes (illustrated schematically, top). 
E. The pixel intensity of Ac-tubulin was quantified in silenced or unsilenced DU145 cells. Cell peripheries were outlined as in C, tubulin intensity integrated 
within the enclosed area, and average intensities in each condition determined. Average intensity values determined from three independent trials (Student’s 
t-test, p¼0.0033). N¼total cell number. 
F,G. DU145 cells expressing control or DIAPH3 shRNAs were stained with Ac-tubulin following staining with rhodamine-phalloidin (F) or FITC-CTxB (G), and 
imaged by fluorescence microscopy. Scale¼10mm (N2 independent trials). 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 751
pEGFR (T669) 
pEGFR (Y992) 
pERK1/2 
1 0.3 
1 1.6 
Mock 
ERK1/2 
8 
pMEK1/2 
(S217/221) 
MEK1/2 
DIAPH3 
-actin 
ERK1/2 
both were phenotypically amoeboid, with numerous membrane 
blebs and rounded morphologies (Fig 8A and B, Supporting 
Information Fig S8A). Enforced DIAPH3 altered the phenotype 
of both cell lines, suppressing amoeboid blebbing and inducing 
formation of prominent lamellipodia, a typical mesenchymal 
feature (Fig 8A, B and F, Supporting Information Fig S8A). 
DIAPH3 also increased levels of the mesenchymal marker 
N-cadherin (Fig 8C), in concert with increased stress fiber 
formation (Fig 8E, Supporting Information Fig S8B), suggesting 
that DIAPH3 promotes an amoeboid to mesenchymal transition. 
The data shown above position DIAPH3 within the EGFR 
pathway. Consistent with this, we identified an EGF-sensitive 
phospho-serine at position 624 in the DIAPH3 primary sequence 
by tandem mass spectrometry (Supporting Information Fig 
S9A). S624 is located within the last of five polyproline/SRC 
homology three binding motifs in the FH1 domain (Fig 8D). This 
site was validated by stable isotope labelling (Supporting 
Control siRNA 
Information Fig S9B and C) and confirmed to be EGF-responsive 
using a custom phosphosite-specific antibody (Supporting 
Information Fig S9D and E). DIAPH3 function was modulated 
by its phosphorylation status at S624. A phospho-null mutant at 
this site (S624A) promoted stress fibre formation, a mesench-ymal 
characteristic (Fig 8E), and suppressed amoeboid blebbing 
(Fig 8F) to a greater degree than the unmodified protein, while a 
phospho-mimetic mutant (S624E) was impaired in both 
activities (Fig 8E and F). These findings support the conclusion 
that DIAPH3 suppresses amoeboid blebbing and is functionally 
inhibited by phosphorylation at S624, which occurs in response 
to EGF treatment (Supporting Information Fig S9E). 
DIAPH3 loss promotes metastasis and is associated with 
metastatic human prostate cancer 
Our findings thus far suggest that DIAPH3 loss promotes motility, 
invasion, and increased oncogenic signalling, all pre-requisites for 
Research Article 
DIAPH3 and the amoeboid phenotype 
pEGFR (Y1068) 
EGFR 
Control 
siRNA 
DIAPH3 
siRNA 
pEGFR (Y1086) 
ERK1/2 
1 4.8 
1 2.0 
DIAPH3 
-actin 
1 4.2 
DIAPH3 
Control 
siRNA 
DIAPH3 
siRNA1 
DIAPH3 
siRNA2 
pERK1/2 
C 
A 
D 10 
pERK/ERK 
(fold of control) 
6 
4 
2 
0 
DIAPH3 siRNA1 
DIAPH3 siRNA2 
B Control 
shRNA 
DIAPH3 
shRNA 
DIAPH3 
100 
80 
60 
40 
20 
0 
Relative 
DIAPH3 level 
(%) 
pERK1/2 
ERK1/2 
Control 
siRNA 
DIAPH3 siRNA 
#1 #2 #3 #4 
E 
Control siRNA 
DIAPH3 siRNA 
50 
40 
30 
20 
10 
0 
0 2 4 6 8 10 
-tubulin 
DIAPH3 
siRNA1 
Control 
siRNA 
EGF (min): 0 2 5 10 0 2 5 10 
pERK1/2 
pERK signal intensity 
(a.u.) 
P=0.0068 
Figure 6. DIAPH3 knockdown enhances EGFR 
signalling. 
A. EGFR is active in DIAPH3-silenced COS7 cells, as 
shown by increased tyrosine phosphorylation at 
activating sites and decreased phosphorylation of 
the T669 inhibitory site. 
B. Enhanced pMEK1/2 in DIAPH3-depleted DU145 
cells. 
C. Targeting of DIAPH3 with four independent siR-NAs 
in COS7 leads to 90% depletion, which 
inversely correlates with pERK1/2 levels. 
D. DIAPH3 depletion in serum-depleted cells 
enhances pERK1/2 levels. 
E. Acute, sustained phosphorylation of ERK1/2 in 
response to EGF in DIAPH3-depleted versus 
control cells (two-way ANOVA, p¼0.0068; N2 
independent trials). 
752  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
80 
60 
40 
HMEC HRASV12 A375P 
100 
80 
60 
40 
20 
100 
80 
60 
40 
20 
0 DIAPH3 shRNA2 
metastasis. Consistentwith this hypothesis, superficial pulmonary 
metastases in nudemice, induced by tail vein injection of DIAPH3- 
silenced DU145 cells, were enhanced relative to unsilenced cells 
(Fig 9A and B). Both the number and size of metastatic foci were 
potentiated by DIAPH3 knockdown (Fig 9B and data not shown). 
Additionally, large tumour thrombotic emboliwere observed only 
in lung sections from mice that underwent injection of DIAPH3- 
silenced cells (Fig 9C). Detection of human cytokeratin 18 (CK18) 
in the lung metastases demonstrated the lesions’ human origin 
(Fig 9D). This was further confirmed by puromycin resistance of 
cells from dissociated lesions in cell culture. Together, these 
findings indicate that DIAPH3 silencing enhances experimental 
metastasis in mice. 
Lastly, we examined expression of the DIAPH3 protein with 
PCa progression using a cohort of 90 human prostate specimens 
in a tissue microarray format and a validated anti-DIAPH3 
antibody (Supporting Information Fig S2B). While DIAPH3 
protein levels did not demonstrably change between benign and 
organ-confined carcinoma, we observed a dramatic reduction of 
DIAPH3 protein levels in metastatic lesions in comparison 
with normal tissue (p¼0.018) and organ-confined tumours 
(p¼0.007, Fig 9E and F). These results are in agreement with 
our genomic analyses (Fig 1E–H) and indicate that loss of the 
DIAPH3 locus functionally results in significant loss of the 
protein in human metastatic disease. 
100 
80 
60 
40 
20 
DISCUSSION 
Research Article 
Martin H. Hager et al. 
Control shRNA 
DIAPH3 shRNA1 
This study provides evidence that the formin DIAPH3 belongs 
to a novel class of metastasis suppressors that inhibits 
conversion to an amoeboid phenotype. Inactivation of 
this gene appears relevant to several human malignancies, 
including prostate and breast carcinomas. We identified a 
consensus area of significant recurrent deletion on chromosome 
13 encompassing the DIAPH3 locus and showed that DIAPH3 
genomic loss and/or decreased DIAPH3 expression occurs 
in organ-confined tumours, but occurs at higher frequency 
in advanced disease, circulating prostate tumour cells, and 
metastatic lesions. We show that DIAPH3 silencing enhances 
tumour cell invasion, promotes amoeboid features in disparate 
cell backgrounds, and enhances experimental metastasis 
in vivo. 
This is the first example of a genomic lesion affecting a direct 
cytoskeletal regulator that governs the amoeboid transition. 
Because amoeboid behaviour enables cells to squeeze through 
gaps in the fibrillar matrix, amoeboid cells are proposed to 
possess a higher proclivity to disseminate and metastasize 
(Sanz-Moreno  Marshall, 2010). We provide evidence that 
DIAPH3 deficiency promotes a wide range of amoeboid 
characteristics, and can cooperate with a canonical activated 
oncogene. In a mammary epithelial background, activated 
20 
DMSO PD98059 DMSO PD98059 DMSO PD98059 
amoeboid cells / field 
(percent of total) 
0 
DU145 
0 
Control EGF EGF + Gefitinib 
amoeboid cells / field 
(percent of total) 
Control shRNA 
DIAPH3 shRNA 
B 
0 
Control EGF EGF + Gefitinib 
DIAPH3 shRNA Control shRNA 
C 
A 
Figure 7. Sensitivity of amoeboid features to MEK1/2 and EGFR inhibition. 
A. Amoeboid features induced by DIAPH3 silencing in human HMEC-HRASV12, A375P melanoma and DU145 cells can be reverted by PD98059 (50 mM). N2 
independent trials. 
B,C. Amoeboid blebbing induced by EGF (10 nM) in control cells is sensitive to the EGFR inhibitor Gefitinib (2 mM), while induction by DIAPH3 silencing is less 
sensitive to both treatments, (C) Representative micrographs, (B) See also Supporting Information Movie S3. 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 753
A PC3 B U87 
DIAPH3 S 
p = 0.0012 
200 
150 
100 
50 
HRAS alone induced EMT; in contrast, DIAPH3 silencing in the 
context of activated HRAS resulted in an amoeboid transition 
consisting of a dramatic morphologic transformation in base-ment 
membrane cultures and high invasive potential (Fig 2). 
These findings suggest that the oncogenic background of tissues 
in which DIAPH3 is lost makes an essential contribution to 
tumour behaviour. 
GFP GFP-DIAPH3 
2.5 p = 0.0048 
GFP 
p = 0.0017 
p = 0.0032 
2.0 
1.5 
1.0 
S624 
20 
15 
10 
5 
We also showed that an endosomal trafficking defect can 
elicit amoeboid behaviour. We propose a model (Fig 10) 
whereby DIAPH3 loss causes cytoskeletal disruption, inhibits 
endocytic down-regulation of RTKs, and leads to persistent 
activation of downstream effectors. That DIAPH3 down-regulation 
enhances endosomal accumulation of EGFR is 
notable given recent reports implicating endocytic trafficking 
Research Article 
DIAPH3 and the amoeboid phenotype 
E Stress fiber formation 
HeLa 
Degree of induction 
(percent of control) 
p  0.0001 
p=0.0291 
F Amoeboid blebbing 
U87 
Membrane blebs per cell 
p  0.0001 
GFP GFP-DIAPH3 
C 
D 
DIAPH3: - + 
N-cadherin 
GFP-DIAPH3 
GFP 
0.5 
-actin 0 
N-cadherin / -actin 
(fold of control) 
GFP-DIAPH3 
GBD DID DD CC FH1 FH2 DAD 
GFP-DIAPH3 variants GFP-DIAPH3 variants 
GFP 
GFP-DIAPH3 
GFP-DIAPH3(S624A) 
GFP-DIAPH3(S624E) 
GFP 
GFP-DIAPH3 
GFP-DIAPH3(S624A) 
GFP-DIAPH3(S624E) 
0 
0 
Figure 8. DIAPH3 expression suppresses the amoeboid phenotype. 
A,B. Micrographs of PC3 (A) and U87 (B) cells expressing GFP or GFP-DIAPH3. Arrows indicate prevalence of amoeboid cells in PC3- or U87-GFP (left panels), and of 
mesenchymal (lamellopodia-enriched) cells in PC3- or U87-GFP-DIAPH3 (right panels). 
C. N-cadherin is significantly upregulated in cells expressing GFP-DIAPH3. 
D. Domain structure of DIAPH3 and location of the S624 phosphosite within the FH1 domain. 
E. Quantitation of stress fiber formation in HeLa cells expressing GFP or DIAPH3 mutants and stained with phalloidin. 
F. Quantitation of amoeboid blebbing in U87 cells expressing GFP or DIAPH3 mutants and stained with CTxB (N2 independent trials). 
754  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
Normal/Benign Carcinoma Metastasis 100 
in tumour suppression (Mosesson et al, 2008) and demonstrat-ing 
increased signalling from EGFR, VEGFR and c-MET if 
endosome processing is compromised (Joffre et al, 2011; 
Lanahan et al, 2010; Wang et al, 2009). Our findings suggest 
that amoeboid behaviour is a disease-relevant outcome of the 
Research Article 
Martin H. Hager et al. 
C Tumour thrombus 
DIAPH3 shRNA 
HE HE 
80 
60 
40 
20 
ability of DIAPH3-silenced tumour cells to usurp the endocytic 
machinery. 
Our model proposes that deregulated endosomal trafficking 
and signalling defects associated with DIAPH3 silencing arise 
from MT disruption. DIAPH3 loss induced MT instability and 
A 
Control shRNA DIAPH3 shRNA 
Control shRNA DIAPH3 shRNA 
Control shRNA 
CK18 CK18 
D 
DIAPH3 shRNA 
Control shRNA 
DIAPH3 shRNA 
p = 0.0493 
Metastases per lung 
B 
Normal/ 
Benign 
PCa Met 
% of samples 
High 
Low 
0 
E F 
Figure 9. Silencing of DIAPH3 promotes metastasis and is associated with metastatic disease in vivo. 
A. DIAPH3-silenced DU145 cells injected into the tail vein of nude mice produced large superficial pulmonary metastases (arrowhead). 
B. Quantification of lung metastases in mice injected with control or DIAPH3-silenced DU145 cells (Mann–Whitney U test). N¼10 mice/condition. 
C. Representative thromboembolus from lung sections from mice injected with DIAPH3-silenced DU145 cells. 
D. Representative lung sections of mice injected with cells expressing control or DIAPH3 shRNAs, stained with HE or an antibody to human CK18. Note presence 
of micrometastases in sections from DIAPH3 shRNA mice (arrowhead; N¼2 independent trials). 
E. DIAPH3 IHC staining of a human tissue microarray (TMA) containing cores with benign human prostate epithelium (Normal/Benign), prostate tumour tissue 
(Carcinoma) and tissue from metastatic lesions (Metastasis). High-power magnifications are shown (bottom). Scale¼200 mm. 
F. DIAPH3 expression is significantly decreased in metastases (Fisher’s exact test, p¼0.018/0.007). 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 755
EGF 
EGFR 
Microtubules 
crotubule 
crotubul 
Amoeboid 
APH3 EGFR-enriched 
EGFR activation, in agreement with reports of a positive 
correlation between tubulin acetylation and EGFR degradation 
(Deribe et al, 2009; Gao et al, 2010). MT instability is emerging 
as a contributor to the amoeboid phenotype (Belletti et al, 2008; 
Berton et al, 2009). Our findings suggest that DIAPH3 is a key 
regulatory node in the transition between amoeboid and 
mesenchymal tumour cell phenotypes and that MT disruption 
may affect amoeboid and mesenchymal features at multiple 
levels. 
The sustained endosomal localization of EGFR consequent to 
DIAPH3 downregulation may be clinically relevant. Response of 
a tumour cell population to a TKI can be influenced by 
distribution of intracellular EGFR, such that some ‘sensitive’ 
cells display EGFR on the plasma membrane while ‘resistant’ 
cells exhibit perinuclear localization (Huang et al, 2009). 
Alternatively, while the ligand-bound receptor undergoes 
normal endocytic trafficking in ‘responsive’ cells, transport to 
lysosomes can be defective in ‘resistant’ cells (Nishimura et al, 
2007). We observed similar responses with DIAPH3 deficiency, 
which markedly reduced sensitivity to a TKI. Our findings are 
suggestive that DIAPH3 loss can induce resistance to EGFR 
inhibitors. This possibility deserves further exploration. 
The RAS/MAPK axis is upregulated in 90% of metastatic PCa 
lesions (Taylor et al, 2010), and hyper-activation of ERK is 
implicated in PCa progression (Gioeli et al, 1999). However, it 
remains unclear how this pathway is activated, since compre-hensive 
profiling of prostate tumours did not reveal activating 
mutations in BRAF or HRAS (Burger et al, 2006; Thomas et al, 
2007). It is notable, then, that in PCa cells DIAPH3 deficiency 
upregulates ERK activity. Our results raise the interesting 
possibility that MEK inhibitors may be useful to target advanced 
disease in patients with tumours with DIAPH3 loss. 
Two recent reports assessed DIAPH3 in the context of cell 
invasion. Lizarraga et al. demonstrated that DIAPH3 silencing 
inhibits formation of filopodia-like invadopodia, invasion and 
degradation of 3D-matrices (Lizarraga et al, 2009). We observed 
DIAPH3 loss to induce a switch to an amoeboid phenotype, in 
which dependence on proteases for invasion is reportedly 
reduced (Friedl  Wolf, 2010). However, while DIAPH3 
silencing suppressed invasion of MDA-MB-231 cells through 
Research Article 
DIAPH3 and the amoeboid phenotype 
Mesenchymal 
Lysosome 
/ 
t 
tubulin heterodimer (disrupted MT) 
Early 
endosome 
DIAPH3: intact 
Focal adhesion maturation 
Stress fiber formation 
DIAPH3: lost 
Lack of focal adhesion maturation 
Cortical actomyosin contractility 
Membrane bleb formation 
Increased motility and invasion 
DIAPH3 
Mic 
1. 
2. 
3. 
2. 
1. 
3. 4. 
5. 
odimer disru 
endosome 
1. 
A 
B 
Figure 10. Model of DIAPH3 perturbation and the 
amoeboid transition. 
A. When DIAPH3 is functional: (1) EGFR is interna-lized 
via endocytosis upon activation; (2) EGFR 
traffics in endosomes along microtubules to 
lysosomes or is recycled back to the plasma 
membrane; (3) ERK activity is attenuated 
following EGFR down-regulation. 
B. When DIAPH3 is lost or inactivated: (1) EGFR is 
internalized. (2) MT are destabilized, preventing 
both transport of EGFR from early endosomes to 
lysosomes, and receptor recycling. (3) Active EGFR 
accumulates in endosomes. (4) ERK activity is 
sustained. (5) Deregulation of proteins promoting 
amoeboid characteristics (e.g. MLC2, FAK) evokes 
cortical actomyosin contraction, focal adhesion 
turnover, and membrane blebbing. Broken 
arrows/lines indicate the presence of signalling 
intermediates. 
756  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
Matrigel (Lizarraga et al, 2009), we observed DIAPH3 silencing 
to promote invasion through collagen I. Using an siRNA screen 
for formin family regulators of membrane blebbing, Stastna 
et al. reported that DIAPH3 silencing inhibited bleb formation 
and promoted cell spreading in HeLa cells (Stastna et al, 2011). 
Of the multiple transcripts of the DIAPH3 locus, Isoform 1 
mediated bleb formation, while an activated variant of Isoform 7 
instead promoted filopodia. Consistent with the last observa-tion, 
we observed a significant reduction in filopodia following 
DIAPH3 silencing of HMEC-HRASV12 (unpublished observa-tions). 
We employed Isoform 7 for our studies, however DIAPH3 
silencing potentiated bleb formation, reduced adhesion and 
increased rates of migration in COS7, DU145 and HMEC. 
Although genetic heterogeneity or different characteristics of 
diverse ECM used in these in vitro studies may play a role in 
these diverse effects, collectively they are consistent with the 
conclusion that DIAPH3 resides at an important signaling node 
that controls invasive behaviour. Importantly, the genomic loss 
data and other findings from human cohorts that we present 
here are consistent with the conclusion that DIAPH3 inactiva-tion 
is likely to promote aggressive behaviour in prostate, breast 
and possibly other tumour types. In the present study, we also 
identified a serine residue in the DIAPH3 FH1 domain 
that seems to result in inactivation of the protein when 
phosphorylated. This finding suggests that DIAPH3 might be 
inactivated by upstream signaling pathways in addition to gene 
disruption. 
Reports have speculated about the presence of a tumour 
suppressor at 13q21 that is independent of RB1. A recent study 
evaluated somatic homozygous deletions (HDs) at high 
resolution in 746 cancer cell lines (Bignell et al, 2010), 
Research Article 
Martin H. Hager et al. 
identifying an ‘unexplained’ HD cluster on chromosome 13 
that exhibits a signature similar to known tumour suppressors. 
This cluster was separate from the dominating HD cluster 
affecting the RB1 locus and had not been assigned to a known 
tumour suppressor gene. DIAPH3 is a candidate non-canonical 
tumour suppressor in this region. 
In conclusion, identification of DIAPH3 as a protein capable 
of mediating the switch between mesenchymal and amoeboid 
phenotypes provides new insight into the molecular processes 
of metastasis, and may facilitate design of more effective 
strategies against advanced disease. 
MATERIALS AND METHODS 
Copy number analysis 
DNA copy number alterations were analysed with the Integrated 
Genomics Viewer using genome-wide GISTIC data (Beroukhim et al, 
2007). DIAPH3 copy number status of PCa patients was analysed using 
comparative genomic hybridization (cCGH) and Affymetrix Genome- 
Wide SNP Array data (Liu et al, 2009). The frequency of DIAPH3 loss in 
circulating tumours cells was assessed using array CGH data (Holcomb 
et al, 2008). 
Immunohistochemistry and tissue microarrays 
The human prostate tissue microarray consisted of normal/benign 
(n¼16), prostate tumour (n¼22) and metastatic tissue cores 
(n ¼24). Immunohistochemistry was performed with DIAPH3 
(HNH3.1) or cytokeratin 18 antibodies. 
Immunoblotting was performed as described in Supporting Informa-tion. 
The paper explained 
PROBLEM: 
While metastatic disease underlies most cancer-related 
mortality, few genetic lesions that select for metastatic tumour 
cell variants have been identified. Amoeboid motility is one of 
several diverse modes adopted by disseminating tumour cells. 
Elucidation of networks and critical signaling nodes that confer 
or restrain the amoeboid phenotype would facilitate discovery of 
novel therapies to control metastasis. The DIAPH3 locus, 
encoding the protein Diaphanous-related formin-3 (DIAPH3), 
resides at a chromosomal location that is frequently lost in 
metastatic prostate cancer. The potential functional significance 
of loss of this locus is unknown. 
RESULTS: 
Analysis of genome-wide, SCNA revealed that the DIAPH3 locus 
was a consensus area of chromosomal deletion common to 
several carcinomas. DIAPH3 deletions accumulated during 
disease progression, were strongly associated with metastatic 
disease, and were prevalent in DTCs from patient bone marrow 
aspirates. DIAPH3 silencing cooperated with oncogenic trans-formation 
to evoke an amoeboid phenotype in several tumour 
cell backgrounds. Loss of DIAPH3 caused cytoskeletal and 
endocytic trafficking defects through which EGFR/MEK/ERK 
signaling was hyperactivated. Pharmacologic inhibition of MEK 
suppressed the amoeboid phenotype, but tyrosine kinase 
inhibitors were ineffective. DIAPH3 silencing potentiated for-mation 
of pulmonary metastases in vivo, and its loss correlated 
with metastasis in human tumours. 
IMPACT: 
This is the first report showing that loss of a cytoskeleton 
remodelling protein, encoded by a locus that is lost at high 
frequency in multiple tumours and is strongly associated with 
metastasis, results in acquisition of the amoeboid cancer cell 
phenotype. These results may have prognostic utility to 
distinguish low-risk from high-risk disease. 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 757
RNAi 
For sequences and transfection methods, see Supporting Information. 
Multiple, independent hairpins and duplexes (4) produced similar 
results in numerous readouts. 
Immunofluorescence 
Antibodies used: Acetylated tubulin and Rab11 (Abcam), FITC-conjugated 
anti-FLAG (Sigma), FITC-conjugated anti-EEA1 (BD Bios-ciences), 
EGFR (Biosource), FITC-Cholera Toxin B (CTxB, Sigma), 
FAK(Y397), MLC2, pMLC2(S19) and Rab5, (Cell Signaling). F-actin 
was detected with rhodamine-phalloidin (Invitrogen). An Axioplan 2 
microscope (Zeiss) equipped with an AxioCam camera was used for 
fluorescence imaging. 
Quantitation of EGFR-positive endosomes and focal adhesions: 
Duplicate slides were evaluated by three observers blinded to the 
experimental group in two independent experiments, in 50 cells/ 
condition per experiment. 
Tubulin acetylation: DU145 cells were incubated with acetylated 
tubulin antibodies, and fluorescence quantified using Axiovision 4.5 
software (Zeiss). Images were rendered using ImageJ processing 
software. Where noted, cells were stained with rhodamine–phalloidin 
or FITC-CTxB prior to fixation. 
Rab11 co-localization: DU145 cells were incubated with antibodies 
against Rab11 and EGFR, and fluorescent co-localization analysed 
with ImageJ. Regions with pixel intensity common to Rab11 and EGFR 
were highlighted. 
Where indicated, cells plated on collagen I were pretreated with 
nocodazole (2mM, Sigma) for 30 min, and processed as above with 
EGFR antibodies. 
Morphogenesis assay 
Three-dimensional culture of epithelial cells was performed as 
described (Debnath et al, 2002). Briefly, cells were seeded onto 
Matrigel basement membrane matrix, and acini size measured and 
analysed with Fiji image processing software. 
Luminescence proliferation assay 
Viable cells were quantified with the CellTiter-Glo assay (Promega), 
with luminescence measured every 24 h with a FLUOstar Omega 
platereader (BMG Labtech). 
Experimental Metastasis Model 
Animal studies were conducted in compliance with Children’s Hospital 
Boston IACUC guidelines using BALB/c nude mice (Massachusetts 
General Hospital). DU145 cells expressing control or DIAPH3 shRNAs 
were resuspended in HBSS, injected into the tail veins of anesthetized 
mice (15 animals per group), and after 8 weeks mice were sacrificed, 
lungs isolated and each lobe fixed individually in 10% formalin. 
Time-lapse video microscopy and track-plot analysis 
Cell migration was monitored with an inverted microscope, collecting 
images every 2 min for 30 h, and movies analysed using Bioimaging 
software (Andor). 
Real-time invasion assay 
Cells labelled with CellTracker Red CMTPX (Molecular Probes) were 
seeded onto collagen I-coated FluoroBlokTM inserts, and fluorescence 
intensity of cells migrated towards chemo-attractants monitored at 6 
or 12 h. 
Statistical analyses 
Student’s t-test (two-tailed) was used if data were normally 
distributed. Otherwise, Mann–Whitney U, ANOVA, or Fisher’s exact 
tests were used. Analyses were performed with Prism 5.0a software. 
Data were plotted as meanSD. 
Author contributions 
MRF, MHH, and SM conceived the study; MHH, SM, DDV and 
MRF designed the experiments; MHH, SM, DDV, MM, KRS and 
JK performed experiments and analysed the data, including 
statistical analyses, with assistance from DRB, SG and GM; WL, 
FD, INH, WBI, HNH and RLV provided reagents, primary data 
sets, and bioinformatics analyses; MHH, SM, and MRF wrote the 
manuscript, with assistance by DDV and RMA. 
Acknowledgements 
The authors thank Joan S. Brugge and Wei Yang for helpful 
discussions, and Delia Lopez, Jiyoung Choi and Maosong Qi for 
technical assistance. Joseph Khoury and Mohini Lutchman 
made contributions in the study’s early phases. Grant support: 
NCI R01 CA143777, CA112303, NIDDK R3747556, P50 
DK65298 and DAMD17-03-2-0033 (M.R.F.); US Department of 
Defense Prostate Cancer Research Program W81XWH-07-1- 
0148 and AUAF/GlaxoSmithKline (M.H.H.); NIH R00 
CA131472 (D.D.V.); American Institute for Cancer Research 
09A107 (S.M.); and NCI R01 CA135008, CA133066 (W.L.). 
Supporting Information is available at EMBO Molecular 
Medicine online. 
The authors declare that they have no conflict of interest. 
References 
Bartolini F, Gundersen GG (2010) Formins and microtubules. Biochim Biophys 
Acta 1803: 164-173 
Bartolini F, Moseley JB, Schmoranzer J, Cassimeris L, Goode BL, Gundersen GG 
(2008) The formin mDia2 stabilizes microtubules independently of its actin 
nucleation activity. J Cell Biol 181: 523-536 
Belletti B, Nicoloso MS, Schiappacassi M, Berton S, Lovat F, Wolf K, Canzonieri 
V, D’Andrea S, Zucchetto A, Friedl P, et al (2008) Stathmin activity influences 
sarcoma cell shape, motility, and metastatic potential. Mol Biol Cell 19: 
2003-2013 
Beroukhim R, Getz G, Nghiemphu L, Barretina J, Hsueh T, Linhart D, Vivanco I, 
Lee JC, Huang JH, Alexander S, et al (2007) Assessing the significance of 
chromosomal aberrations in cancer: Methodology and application to 
glioma. Proc Natl Acad Sci USA 104: 20007-20012 
Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, 
Barretina J, Boehm JS, Dobson J, Urashima M, et al (2010) The landscape of 
somatic copy-number alteration across human cancers. Nature 463: 899- 
905 
Berton S, Belletti B, Wolf K, Canzonieri V, Lovat F, Vecchione A, Colombatti A, 
Friedl P, Baldassarre G (2009) The tumor suppressor functions of p27(kip1) 
include control of the mesenchymal/amoeboid transition. Mol Cell Biol 29: 
5031-5045 
Research Article 
DIAPH3 and the amoeboid phenotype 
758  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
Bignell GR, Greenman CD, Davies H, Butler AP, Edkins S, Andrews JM, Buck G, 
Chen L, Beare D, Latimer C, et al (2010) Signatures of mutation and selection 
in the cancer genome. Nature 463: 893-898 
Burger M, Denzinger S, Hammerschmied C, Tannapfel A, Maderstorfer A, 
Wieland WF, Hartmann A, Stoehr R (2006) Mitogen-activated protein kinase 
signaling is activated in prostate tumors but not mediated by B-RAF 
mutations. Eur Urol 50: 1102-1109; discussion 1109–1110 
Chalkia D, Nikolaidis N, Makalowski W, Klein J, Nei M (2008) Origins and 
evolution of the formin multigene family that is involved in the formation of 
actin filaments. Mol Biol Evol 25: 2717-2733 
Debnath J, Mills KR, Collins NL, Reginato MJ, Muthuswamy SK, Brugge JS 
(2002) The role of apoptosis in creating and maintaining luminal space 
within normal and oncogene-expressing mammary acini. Cell 111: 
29-40 
Deribe YL, Wild P, Chandrashaker A, Curak J, Schmidt MH, Kalaidzidis Y, 
Milutinovic N, Kratchmarova I, Buerkle L, Fetchko MJ, et al (2009) 
Regulation of epidermal growth factor receptor trafficking by lysine 
deacetylase HDAC6. Sci Signal 2: ra84 
Di Vizio D, Kim J, Hager MH, Morello M, Yang W, Lafargue CJ, True LD, Rubin 
MA, Adam RM, Beroukhim R, et al (2009) Oncosome formation in prostate 
cancer: Association with a region of frequent chromosomal deletion in 
metastatic disease. Cancer Res 69: 5601-5609 
Dong JT, Chen C, Stultz BG, Isaacs JT, Frierson HF, Jr (2000) Deletion at 13q21 is 
associated with aggressive prostate cancers. Cancer Res 60: 3880- 
3883 
Fernandez-Borja M, Janssen L, Verwoerd D, Hordijk P, Neefjes J (2005) RhoB 
regulates endosome transport by promoting actin assembly on endosomal 
membranes through Dia1. J Cell Sci 118: 2661-2670 
Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, Chen H, 
Omeroglu G, Meterissian S, Omeroglu A, et al (2008) Stromal gene 
expression predicts clinical outcome in breast cancer. Nat Med 14: 518- 
527 
Friedl P, Wolf K (2010) Plasticity of cell migration: A multiscale tuning model. J 
Cell Biol 188: 11-19 
Gaillard J, Ramabhadran V, Neumann E, Gurel P, Blanchoin L, Vantard M, Higgs 
HN (2011) Differential interactions of the formins INF2, mDia1, and mDia2 
with microtubules. Mol Biol Cell 22: 4575-4587 
Gao YS, Hubbert CC, Yao TP (2010) The microtubule-associated histone 
deacetylase 6 (HDAC6) regulates epidermal growth factor receptor (EGFR) 
endocytic trafficking and degradation. J Biol Chem 285: 11219-11226 
Gasman S, Kalaidzidis Y, Zerial M (2003) RhoD regulates endosome dynamics 
through diaphanous-related formin and Src tyrosine kinase. Nat Cell Biol 5: 
195-204 
Gioeli D, Mandell JW, Petroni GR, Frierson HF, Jr, WeberMJ (1999) Activation of 
mitogen-activated protein kinase associated with prostate cancer 
progression. Cancer Res 59: 279-284 
Goode BL, Eck MJ (2007) Mechanism and function of formins in the control of 
actin assembly. Annu Rev Biochem 76: 593-627 
Hamadi A, Bouali M, Dontenwill M, Stoeckel H, Takeda K, Ronde P (2005) 
Regulation of focal adhesion dynamics and disassembly by phosphorylation 
of FAK at tyrosine 397. J Cell Sci 118: 4415-4425 
Holcomb IN, Grove DI, Kinnunen M, Friedman CL, Gallaher IS, Morgan TM, 
Sather CL, Delrow JJ, Nelson PS, Lange PH, et al (2008) Genomic alterations 
indicate tumor origin and varied metastatic potential of disseminated cells 
from prostate cancer patients. Cancer Res 68: 5599-5608 
Hosoki S, Ota S, Ichikawa Y, Suzuki H, Ueda T, Naya Y, Akakura K, Igarashi T, 
Oshimura M, Nihei N, et al (2002) Suppression of metastasis of rat prostate 
cancer by introduction of human chromosome 13. Asian J Androl 4: 131- 
136 
Huang DH, Su L, Peng XH, Zhang H, Khuri FR, Shin DM, Chen ZG (2009) 
Quantum dot-based quantification revealed differences in subcellular 
localization of EGFR and E-cadherin between EGFR-TKI sensitive and 
insensitive cancer cells. Nanotechnology 20: 225102 
Joffre C, Barrow R, Menard L, Calleja V, Hart IR, Kermorgant S (2011) A direct 
role for Met endocytosis in tumorigenesis. Nat Cell Biol 13: 827-837 
Research Article 
Martin H. Hager et al. 
Kainu T, Juo SH, Desper R, Schaffer AA, Gillanders E, Rozenblum E, Freas-Lutz D, 
Weaver D, Stephan D, Bailey-Wilson J, et al (2000) Somatic deletions in 
hereditary breast cancers implicate 13q21 as a putative novel breast cancer 
susceptibility locus. Proc Natl Acad Sci USA 97: 9603-9608 
Kalluri R, Weinberg RA (2009) The basics of epithelial–mesenchymal 
transition. J Clin Invest 119: 1420-1428 
Lanahan AA, Hermans K, Claes F, Kerley-Hamilton JS, Zhuang ZW, Giordano FJ, 
Carmeliet P, Simons M (2010) VEGF receptor 2 endocytic trafficking 
regulates arterial morphogenesis. Dev Cell 18: 713-724 
Li HR, Wang-Rodriguez J, Nair TM, Yeakley JM, Kwon YS, Bibikova M, Zheng C, 
Zhou L, Zhang K, Downs T et al (2006) Two-dimensional transcriptome 
profiling: Identification of messenger RNA isoform signatures in prostate 
cancer from archived paraffin-embedded cancer specimens. Cancer Res 66: 
4079-4088 
Liu W, Laitinen S, Khan S, Vihinen M, Kowalski J, Yu G, Chen L, Ewing CM, 
Eisenberger MA, Carducci MA, et al (2009) Copy number analysis indicates 
monoclonal origin of lethal metastatic prostate cancer. Nat Med 15: 559- 
565 
Lizarraga F, Poincloux R, Romao M, Montagnac G, Le Dez G, Bonne I, Rigaill G, 
Raposo G, Chavrier P (2009) Diaphanous-related formins are required for 
invadopodia formation and invasion of breast tumor cells. Cancer Res 69: 
2792-2800 
Mas VR, Maluf DG, Archer KJ, Yanek K, Kong X, Kulik L, Freise CE, Olthoff KM, 
Ghobrial RM, McIver P, et al (2009) Genes involved in viral carcinogenesis 
and tumor initiation in hepatitis C virus-induced hepatocellular carcinoma. 
Mol Med 15: 85-94 
Mosesson Y, Mills GB, Yarden Y (2008) Derailed endocytosis: An emerging 
feature of cancer. Nat Rev Cancer 8: 835-850 
Nishimura Y, Bereczky B, Ono M (2007) The EGFR inhibitor gefitinib 
suppresses ligand-stimulated endocytosis of EGFR via the early/late 
endocytic pathway in non-small cell lung cancer cell lines. Histochem Cell 
Biol 127: 541-553 
Peng J, Wallar BJ, Flanders A, Swiatek PJ, Alberts AS (2003) Disruption of the 
Diaphanous-related formin Drf1 gene encoding mDia1 reveals a role for 
Drf3 as an effector for Cdc42. Curr Biol 13: 534-545 
Pinner S, Sahai E (2008) PDK1 regulates cancer cell motility by antagonising 
inhibition of ROCK1 by RhoE. Nat Cell Biol 10: 127-137 
Sahai E, Marshall CJ (2003) Differing modes of tumour cell invasion have 
distinct requirements for Rho/ROCK signalling and extracellular proteolysis. 
Nat Cell Biol 5: 711-719 
Sanz-Moreno V, Gadea G, Ahn J, Paterson H, Marra P, Pinner S, Sahai E, 
Marshall CJ (2008) Rac activation and inactivation control plasticity of 
tumor cell movement. Cell 135: 510-523 
Sanz-Moreno V, Marshall CJ (2010) The plasticity of cytoskeletal dynamics 
underlying neoplastic cell migration. Curr Opin Cell Biol 22: 690-696 
Schmidt S, Friedl P (2010) Interstitial cell migration: Integrin-dependent and 
alternative adhesion mechanisms. Cell Tissue Res 339: 83-92 
Soldati T, Schliwa M (2006) Powering membrane traffic in endocytosis and 
recycling. Nat Rev Mol Cell Biol 7: 897-908 
Sonee M, Barron E, Yarber FA, Hamm-Alvarez SF (1998) Taxol inhibits 
endosomal–lysosomal membrane trafficking at two distinct steps in CV-1 
cells. Am J Physiol 275: C1630-C1639 
Sorkin A, von Zastrow M (2009) Endocytosis and signalling: Intertwining 
molecular networks. Nat Rev Mol Cell Biol 10: 609-622 
Stastna J, Pan X, Wang H, Kollmannsperger A, Kutscheidt S, Lohmann V, Grosse 
R, Fackler OT (2011) Differing and isoform-specific roles for the formin 
DIAPH3 in plasma membrane blebbing and filopodia formation. Cell Res 22: 
728-745 
Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, 
Kaushik P, Cerami E, Reva B, et al (2010) Integrative genomic profiling of 
human prostate cancer. Cancer Cell 18: 11-22 
Thomas RK, Baker AC, Debiasi RM, Winckler W, Laframboise T, Lin WM, Wang 
M, Feng W, Zander T, MacConaill L, et al (2007) High-throughput oncogene 
mutation profiling in human cancer. Nat Genet 39: 347-351 
Verhey KJ, Gaertig J (2007) The tubulin code. Cell Cycle 6: 2152-2160 
www.embomolmed.org EMBO Mol Med 4, 743–760  2012 EMBO Molecular Medicine 759
Wallar BJ, Deward AD, Resau JH, Alberts AS (2007) RhoB and the mammalian 
Diaphanous-related formin mDia2 in endosome trafficking. Exp Cell Res 
313: 560-571 
Wang Y, Roche O, Yan MS, Finak G, Evans AJ, Metcalf JL, Hast BE, Hanna SC, 
Wondergem B, Furge KA, et al (2009) Regulation of endocytosis via the 
oxygen-sensing pathway. Nat Med 15: 319-324 
Wikman H, Vessella R, Pantel K (2008) Cancer micrometastasis and tumour 
dormancy. Apmis 116: 754-770 
Wolf K, Mazo I, Leung H, Engelke K, von Andrian UH, Deryugina EI, Strongin AY, 
Brocker EB, Friedl P (2003) Compensation mechanism in tumor cell 
migration: Mesenchymal–amoeboid transition after blocking of pericellular 
proteolysis. J Cell Biol 160: 267-277 
Wyckoff JB, Pinner SE, Gschmeissner S, Condeelis JS, Sahai E (2006) ROCK- and 
myosin-dependent matrix deformation enables protease-independent 
tumor–cell invasion in vivo. Curr Biol 16: 1515-1523 
Research Article 
DIAPH3 and the amoeboid phenotype 
760  2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org

More Related Content

What's hot

Jenny Giannopoulou, Prostate cancer methylome, fged_seattle_2013
Jenny Giannopoulou, Prostate cancer methylome, fged_seattle_2013Jenny Giannopoulou, Prostate cancer methylome, fged_seattle_2013
Jenny Giannopoulou, Prostate cancer methylome, fged_seattle_2013Functional Genomics Data Society
 
Differences in microRNA expression during tumor development in the transition...
Differences in microRNA expression during tumor development in the transition...Differences in microRNA expression during tumor development in the transition...
Differences in microRNA expression during tumor development in the transition...Enrique Moreno Gonzalez
 
Molecular Genetics of Cancer
Molecular Genetics of CancerMolecular Genetics of Cancer
Molecular Genetics of CancerNeha Vats
 
miRNA Breast Cancer Prognosis -- Ingenuity Systems
miRNA Breast Cancer Prognosis -- Ingenuity SystemsmiRNA Breast Cancer Prognosis -- Ingenuity Systems
miRNA Breast Cancer Prognosis -- Ingenuity SystemsNatalie Ng
 
Melanoma frontline approach
Melanoma frontline approachMelanoma frontline approach
Melanoma frontline approachEreny Samwel
 
Cancer prevention and treatment
Cancer prevention and treatmentCancer prevention and treatment
Cancer prevention and treatmentTze Hua Yeu
 
Microsatellite instability pathway and molecular genetics of colorectal cancer
Microsatellite instability pathway and molecular genetics of colorectal cancerMicrosatellite instability pathway and molecular genetics of colorectal cancer
Microsatellite instability pathway and molecular genetics of colorectal cancerDr.Juel Chowdhury MD
 
Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael Araya
 
The most complete map of oncogenes to date a summary of 568 oncogenes in 66 c...
The most complete map of oncogenes to date a summary of 568 oncogenes in 66 c...The most complete map of oncogenes to date a summary of 568 oncogenes in 66 c...
The most complete map of oncogenes to date a summary of 568 oncogenes in 66 c...DoriaFang
 
Cancer by online teaching ot
Cancer by online teaching otCancer by online teaching ot
Cancer by online teaching otvidan biology
 
GTC Group 4 Assignment
GTC Group 4 AssignmentGTC Group 4 Assignment
GTC Group 4 AssignmentCalvin Chan
 
Cancer therapy at protein level
Cancer therapy at protein levelCancer therapy at protein level
Cancer therapy at protein levelSuganyaPaulraj
 
Cancer therapy at gene level
Cancer therapy at gene levelCancer therapy at gene level
Cancer therapy at gene levelSuganyaPaulraj
 
A germline mutation in the brca1 3'utr predicts stage iv breast cancer
A germline mutation in the brca1 3'utr predicts stage iv breast cancerA germline mutation in the brca1 3'utr predicts stage iv breast cancer
A germline mutation in the brca1 3'utr predicts stage iv breast cancerDavid W. Salzman
 
X linked diseases-mitochondrial diseases
X linked diseases-mitochondrial diseasesX linked diseases-mitochondrial diseases
X linked diseases-mitochondrial diseasesdr_ekbalabohashem
 

What's hot (20)

Jenny Giannopoulou, Prostate cancer methylome, fged_seattle_2013
Jenny Giannopoulou, Prostate cancer methylome, fged_seattle_2013Jenny Giannopoulou, Prostate cancer methylome, fged_seattle_2013
Jenny Giannopoulou, Prostate cancer methylome, fged_seattle_2013
 
Differences in microRNA expression during tumor development in the transition...
Differences in microRNA expression during tumor development in the transition...Differences in microRNA expression during tumor development in the transition...
Differences in microRNA expression during tumor development in the transition...
 
Molecular Genetics of Cancer
Molecular Genetics of CancerMolecular Genetics of Cancer
Molecular Genetics of Cancer
 
miRNA Breast Cancer Prognosis -- Ingenuity Systems
miRNA Breast Cancer Prognosis -- Ingenuity SystemsmiRNA Breast Cancer Prognosis -- Ingenuity Systems
miRNA Breast Cancer Prognosis -- Ingenuity Systems
 
Melanoma frontline approach
Melanoma frontline approachMelanoma frontline approach
Melanoma frontline approach
 
Cancer prevention and treatment
Cancer prevention and treatmentCancer prevention and treatment
Cancer prevention and treatment
 
Gtc presentation 2
Gtc presentation 2Gtc presentation 2
Gtc presentation 2
 
Research Poster
Research PosterResearch Poster
Research Poster
 
Microsatellite instability pathway and molecular genetics of colorectal cancer
Microsatellite instability pathway and molecular genetics of colorectal cancerMicrosatellite instability pathway and molecular genetics of colorectal cancer
Microsatellite instability pathway and molecular genetics of colorectal cancer
 
Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW edits
 
The most complete map of oncogenes to date a summary of 568 oncogenes in 66 c...
The most complete map of oncogenes to date a summary of 568 oncogenes in 66 c...The most complete map of oncogenes to date a summary of 568 oncogenes in 66 c...
The most complete map of oncogenes to date a summary of 568 oncogenes in 66 c...
 
Carcinogenesis
CarcinogenesisCarcinogenesis
Carcinogenesis
 
Pharmacogenomics mine
Pharmacogenomics minePharmacogenomics mine
Pharmacogenomics mine
 
Cancer by online teaching ot
Cancer by online teaching otCancer by online teaching ot
Cancer by online teaching ot
 
GTC Group 4 Assignment
GTC Group 4 AssignmentGTC Group 4 Assignment
GTC Group 4 Assignment
 
Cancer therapy at protein level
Cancer therapy at protein levelCancer therapy at protein level
Cancer therapy at protein level
 
Seminar mol biol_1_spring_2013
Seminar mol biol_1_spring_2013Seminar mol biol_1_spring_2013
Seminar mol biol_1_spring_2013
 
Cancer therapy at gene level
Cancer therapy at gene levelCancer therapy at gene level
Cancer therapy at gene level
 
A germline mutation in the brca1 3'utr predicts stage iv breast cancer
A germline mutation in the brca1 3'utr predicts stage iv breast cancerA germline mutation in the brca1 3'utr predicts stage iv breast cancer
A germline mutation in the brca1 3'utr predicts stage iv breast cancer
 
X linked diseases-mitochondrial diseases
X linked diseases-mitochondrial diseasesX linked diseases-mitochondrial diseases
X linked diseases-mitochondrial diseases
 

Similar to Dr. Hager Scholarship at Harvard (2012)

Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...
Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...
Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...Enrique Moreno Gonzalez
 
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...Sehrish Rafique
 
3478-43782-3-PB
3478-43782-3-PB3478-43782-3-PB
3478-43782-3-PB东妮 郑
 
Comparison of risk factors and molecular analysis of right-sided colon and le...
Comparison of risk factors and molecular analysis of right-sided colon and le...Comparison of risk factors and molecular analysis of right-sided colon and le...
Comparison of risk factors and molecular analysis of right-sided colon and le...M. Luisetto Pharm.D.Spec. Pharmacology
 
The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...Journal of Research in Biology
 
The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...Journal of Research in Biology
 
Kurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLSKurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLSSwati Jalgaonkar
 
BRCA1 and BRCA 2 breast cancer.pdf
BRCA1 and BRCA 2 breast cancer.pdfBRCA1 and BRCA 2 breast cancer.pdf
BRCA1 and BRCA 2 breast cancer.pdfedisoneta
 
International Journal of Biometrics and Bioinformatics(IJBB) Volume (3) Issue...
International Journal of Biometrics and Bioinformatics(IJBB) Volume (3) Issue...International Journal of Biometrics and Bioinformatics(IJBB) Volume (3) Issue...
International Journal of Biometrics and Bioinformatics(IJBB) Volume (3) Issue...CSCJournals
 
Alain Toledano : Test and genomic profile : what future in breast cancer trea...
Alain Toledano : Test and genomic profile : what future in breast cancer trea...Alain Toledano : Test and genomic profile : what future in breast cancer trea...
Alain Toledano : Test and genomic profile : what future in breast cancer trea...breastcancerupdatecongress
 
Cancer Res-2015-Bonastre-1287-97
Cancer Res-2015-Bonastre-1287-97Cancer Res-2015-Bonastre-1287-97
Cancer Res-2015-Bonastre-1287-97Sara Verdura
 
biochem of cancer modified dialysis treatment
biochem of cancer modified dialysis treatmentbiochem of cancer modified dialysis treatment
biochem of cancer modified dialysis treatmentThomas Brinkman
 
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docxCell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docxzebadiahsummers
 
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docxCell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docxketurahhazelhurst
 

Similar to Dr. Hager Scholarship at Harvard (2012) (20)

Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...
Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...
Odontogenic ameloblast-associated protein (ODAM) inhibits growth and migratio...
 
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
Estrogen-induced chromatin decondensation and nuclear re-organization linked ...
 
poster_Xingzhi2
poster_Xingzhi2poster_Xingzhi2
poster_Xingzhi2
 
news and views
news and viewsnews and views
news and views
 
ACC Cancer Cell May 2016
ACC Cancer Cell May 2016ACC Cancer Cell May 2016
ACC Cancer Cell May 2016
 
3478-43782-3-PB
3478-43782-3-PB3478-43782-3-PB
3478-43782-3-PB
 
Comparison of risk factors and molecular analysis of right-sided colon and le...
Comparison of risk factors and molecular analysis of right-sided colon and le...Comparison of risk factors and molecular analysis of right-sided colon and le...
Comparison of risk factors and molecular analysis of right-sided colon and le...
 
The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...
 
The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...
 
CRC CHP final PDF
CRC CHP final PDFCRC CHP final PDF
CRC CHP final PDF
 
Kurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLSKurrey_et_al-2009-STEM_CELLS
Kurrey_et_al-2009-STEM_CELLS
 
MasterThesis
MasterThesisMasterThesis
MasterThesis
 
BRCA1 and BRCA 2 breast cancer.pdf
BRCA1 and BRCA 2 breast cancer.pdfBRCA1 and BRCA 2 breast cancer.pdf
BRCA1 and BRCA 2 breast cancer.pdf
 
International Journal of Biometrics and Bioinformatics(IJBB) Volume (3) Issue...
International Journal of Biometrics and Bioinformatics(IJBB) Volume (3) Issue...International Journal of Biometrics and Bioinformatics(IJBB) Volume (3) Issue...
International Journal of Biometrics and Bioinformatics(IJBB) Volume (3) Issue...
 
Alain Toledano : Test and genomic profile : what future in breast cancer trea...
Alain Toledano : Test and genomic profile : what future in breast cancer trea...Alain Toledano : Test and genomic profile : what future in breast cancer trea...
Alain Toledano : Test and genomic profile : what future in breast cancer trea...
 
Cancer Res-2015-Bonastre-1287-97
Cancer Res-2015-Bonastre-1287-97Cancer Res-2015-Bonastre-1287-97
Cancer Res-2015-Bonastre-1287-97
 
biochem of cancer modified dialysis treatment
biochem of cancer modified dialysis treatmentbiochem of cancer modified dialysis treatment
biochem of cancer modified dialysis treatment
 
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docxCell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
 
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docxCell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cel.docx
 
Report CTX 2015
Report CTX 2015Report CTX 2015
Report CTX 2015
 

More from Dr. Martin Hager, MBA

Dr. Hager 2022 Interview Gesundhyte.de Magazine, page 29
Dr. Hager 2022 Interview Gesundhyte.de Magazine, page 29Dr. Hager 2022 Interview Gesundhyte.de Magazine, page 29
Dr. Hager 2022 Interview Gesundhyte.de Magazine, page 29Dr. Martin Hager, MBA
 
Dr. Hager 2021 Interview Roche in Deutschland (page 3)
Dr. Hager 2021 Interview Roche in Deutschland (page 3)Dr. Hager 2021 Interview Roche in Deutschland (page 3)
Dr. Hager 2021 Interview Roche in Deutschland (page 3)Dr. Martin Hager, MBA
 
Dr. Hager 2020 Interview Journal Onkologie
Dr. Hager 2020 Interview Journal OnkologieDr. Hager 2020 Interview Journal Onkologie
Dr. Hager 2020 Interview Journal OnkologieDr. Martin Hager, MBA
 
Dr. Hager 2020 Interview VISION (page 21)
Dr. Hager 2020 Interview VISION (page 21)Dr. Hager 2020 Interview VISION (page 21)
Dr. Hager 2020 Interview VISION (page 21)Dr. Martin Hager, MBA
 
Dr. Hager 2020 Interview Frankfurter Allgemeine Zeitung
Dr. Hager 2020 Interview Frankfurter Allgemeine ZeitungDr. Hager 2020 Interview Frankfurter Allgemeine Zeitung
Dr. Hager 2020 Interview Frankfurter Allgemeine ZeitungDr. Martin Hager, MBA
 
Dr. Hager 2019 Interview Bilanz Das Deutsche Wirtschaftsmagazin
Dr. Hager 2019 Interview Bilanz Das Deutsche WirtschaftsmagazinDr. Hager 2019 Interview Bilanz Das Deutsche Wirtschaftsmagazin
Dr. Hager 2019 Interview Bilanz Das Deutsche WirtschaftsmagazinDr. Martin Hager, MBA
 
Dr. Hager 2016 Presentation The Challenges of Achieving Early Efficacy in Cli...
Dr. Hager 2016 Presentation The Challenges of Achieving Early Efficacy in Cli...Dr. Hager 2016 Presentation The Challenges of Achieving Early Efficacy in Cli...
Dr. Hager 2016 Presentation The Challenges of Achieving Early Efficacy in Cli...Dr. Martin Hager, MBA
 
Dr. Hager 2018 presentation Roche media event ESMO
Dr. Hager 2018 presentation Roche media event ESMODr. Hager 2018 presentation Roche media event ESMO
Dr. Hager 2018 presentation Roche media event ESMODr. Martin Hager, MBA
 
Dr. Hager 2018 presentation: Roche PHC 2.0 press conference
Dr. Hager 2018 presentation: Roche PHC 2.0 press conferenceDr. Hager 2018 presentation: Roche PHC 2.0 press conference
Dr. Hager 2018 presentation: Roche PHC 2.0 press conferenceDr. Martin Hager, MBA
 
Dr. Hager 2018 Interview "Im Focus Onkologie"
Dr. Hager 2018 Interview "Im Focus Onkologie"Dr. Hager 2018 Interview "Im Focus Onkologie"
Dr. Hager 2018 Interview "Im Focus Onkologie"Dr. Martin Hager, MBA
 
MBA Practicum: Global Venture Capital
MBA Practicum: Global Venture CapitalMBA Practicum: Global Venture Capital
MBA Practicum: Global Venture CapitalDr. Martin Hager, MBA
 
Dr. Hager 2014 interview, Daiichi Sankyo Co. Ltd. (page 64)
Dr. Hager 2014 interview, Daiichi Sankyo Co. Ltd. (page 64)Dr. Hager 2014 interview, Daiichi Sankyo Co. Ltd. (page 64)
Dr. Hager 2014 interview, Daiichi Sankyo Co. Ltd. (page 64)Dr. Martin Hager, MBA
 
Dr. Hager 2012 interview Daiichi Sankyo Co. Ltd.
Dr. Hager 2012 interview Daiichi Sankyo Co. Ltd. Dr. Hager 2012 interview Daiichi Sankyo Co. Ltd.
Dr. Hager 2012 interview Daiichi Sankyo Co. Ltd. Dr. Martin Hager, MBA
 

More from Dr. Martin Hager, MBA (14)

Dr. Hager 2022 Interview Gesundhyte.de Magazine, page 29
Dr. Hager 2022 Interview Gesundhyte.de Magazine, page 29Dr. Hager 2022 Interview Gesundhyte.de Magazine, page 29
Dr. Hager 2022 Interview Gesundhyte.de Magazine, page 29
 
Dr. Hager 2021 Interview Roche in Deutschland (page 3)
Dr. Hager 2021 Interview Roche in Deutschland (page 3)Dr. Hager 2021 Interview Roche in Deutschland (page 3)
Dr. Hager 2021 Interview Roche in Deutschland (page 3)
 
Dr. Hager 2021 Interview
Dr. Hager 2021 Interview Dr. Hager 2021 Interview
Dr. Hager 2021 Interview
 
Dr. Hager 2020 Interview Journal Onkologie
Dr. Hager 2020 Interview Journal OnkologieDr. Hager 2020 Interview Journal Onkologie
Dr. Hager 2020 Interview Journal Onkologie
 
Dr. Hager 2020 Interview VISION (page 21)
Dr. Hager 2020 Interview VISION (page 21)Dr. Hager 2020 Interview VISION (page 21)
Dr. Hager 2020 Interview VISION (page 21)
 
Dr. Hager 2020 Interview Frankfurter Allgemeine Zeitung
Dr. Hager 2020 Interview Frankfurter Allgemeine ZeitungDr. Hager 2020 Interview Frankfurter Allgemeine Zeitung
Dr. Hager 2020 Interview Frankfurter Allgemeine Zeitung
 
Dr. Hager 2019 Interview Bilanz Das Deutsche Wirtschaftsmagazin
Dr. Hager 2019 Interview Bilanz Das Deutsche WirtschaftsmagazinDr. Hager 2019 Interview Bilanz Das Deutsche Wirtschaftsmagazin
Dr. Hager 2019 Interview Bilanz Das Deutsche Wirtschaftsmagazin
 
Dr. Hager 2016 Presentation The Challenges of Achieving Early Efficacy in Cli...
Dr. Hager 2016 Presentation The Challenges of Achieving Early Efficacy in Cli...Dr. Hager 2016 Presentation The Challenges of Achieving Early Efficacy in Cli...
Dr. Hager 2016 Presentation The Challenges of Achieving Early Efficacy in Cli...
 
Dr. Hager 2018 presentation Roche media event ESMO
Dr. Hager 2018 presentation Roche media event ESMODr. Hager 2018 presentation Roche media event ESMO
Dr. Hager 2018 presentation Roche media event ESMO
 
Dr. Hager 2018 presentation: Roche PHC 2.0 press conference
Dr. Hager 2018 presentation: Roche PHC 2.0 press conferenceDr. Hager 2018 presentation: Roche PHC 2.0 press conference
Dr. Hager 2018 presentation: Roche PHC 2.0 press conference
 
Dr. Hager 2018 Interview "Im Focus Onkologie"
Dr. Hager 2018 Interview "Im Focus Onkologie"Dr. Hager 2018 Interview "Im Focus Onkologie"
Dr. Hager 2018 Interview "Im Focus Onkologie"
 
MBA Practicum: Global Venture Capital
MBA Practicum: Global Venture CapitalMBA Practicum: Global Venture Capital
MBA Practicum: Global Venture Capital
 
Dr. Hager 2014 interview, Daiichi Sankyo Co. Ltd. (page 64)
Dr. Hager 2014 interview, Daiichi Sankyo Co. Ltd. (page 64)Dr. Hager 2014 interview, Daiichi Sankyo Co. Ltd. (page 64)
Dr. Hager 2014 interview, Daiichi Sankyo Co. Ltd. (page 64)
 
Dr. Hager 2012 interview Daiichi Sankyo Co. Ltd.
Dr. Hager 2012 interview Daiichi Sankyo Co. Ltd. Dr. Hager 2012 interview Daiichi Sankyo Co. Ltd.
Dr. Hager 2012 interview Daiichi Sankyo Co. Ltd.
 

Recently uploaded

Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝soniya singh
 
BREEDING FOR RESISTANCE TO BIOTIC STRESS.pptx
BREEDING FOR RESISTANCE TO BIOTIC STRESS.pptxBREEDING FOR RESISTANCE TO BIOTIC STRESS.pptx
BREEDING FOR RESISTANCE TO BIOTIC STRESS.pptxPABOLU TEJASREE
 
Vision and reflection on Mining Software Repositories research in 2024
Vision and reflection on Mining Software Repositories research in 2024Vision and reflection on Mining Software Repositories research in 2024
Vision and reflection on Mining Software Repositories research in 2024AyushiRastogi48
 
Call Girls In Nihal Vihar Delhi ❤️8860477959 Looking Escorts In 24/7 Delhi NCR
Call Girls In Nihal Vihar Delhi ❤️8860477959 Looking Escorts In 24/7 Delhi NCRCall Girls In Nihal Vihar Delhi ❤️8860477959 Looking Escorts In 24/7 Delhi NCR
Call Girls In Nihal Vihar Delhi ❤️8860477959 Looking Escorts In 24/7 Delhi NCRlizamodels9
 
Scheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxScheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxyaramohamed343013
 
Transposable elements in prokaryotes.ppt
Transposable elements in prokaryotes.pptTransposable elements in prokaryotes.ppt
Transposable elements in prokaryotes.pptArshadWarsi13
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfSwapnil Therkar
 
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxTHE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxNandakishor Bhaurao Deshmukh
 
OECD bibliometric indicators: Selected highlights, April 2024
OECD bibliometric indicators: Selected highlights, April 2024OECD bibliometric indicators: Selected highlights, April 2024
OECD bibliometric indicators: Selected highlights, April 2024innovationoecd
 
BIOETHICS IN RECOMBINANT DNA TECHNOLOGY.
BIOETHICS IN RECOMBINANT DNA TECHNOLOGY.BIOETHICS IN RECOMBINANT DNA TECHNOLOGY.
BIOETHICS IN RECOMBINANT DNA TECHNOLOGY.PraveenaKalaiselvan1
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real timeSatoshi NAKAHIRA
 
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...lizamodels9
 
(9818099198) Call Girls In Noida Sector 14 (NOIDA ESCORTS)
(9818099198) Call Girls In Noida Sector 14 (NOIDA ESCORTS)(9818099198) Call Girls In Noida Sector 14 (NOIDA ESCORTS)
(9818099198) Call Girls In Noida Sector 14 (NOIDA ESCORTS)riyaescorts54
 
Evidences of Evolution General Biology 2
Evidences of Evolution General Biology 2Evidences of Evolution General Biology 2
Evidences of Evolution General Biology 2John Carlo Rollon
 
Call Us ≽ 9953322196 ≼ Call Girls In Lajpat Nagar (Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Lajpat Nagar (Delhi) |Call Us ≽ 9953322196 ≼ Call Girls In Lajpat Nagar (Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Lajpat Nagar (Delhi) |aasikanpl
 
Artificial Intelligence In Microbiology by Dr. Prince C P
Artificial Intelligence In Microbiology by Dr. Prince C PArtificial Intelligence In Microbiology by Dr. Prince C P
Artificial Intelligence In Microbiology by Dr. Prince C PPRINCE C P
 
Neurodevelopmental disorders according to the dsm 5 tr
Neurodevelopmental disorders according to the dsm 5 trNeurodevelopmental disorders according to the dsm 5 tr
Neurodevelopmental disorders according to the dsm 5 trssuser06f238
 
GenBio2 - Lesson 1 - Introduction to Genetics.pptx
GenBio2 - Lesson 1 - Introduction to Genetics.pptxGenBio2 - Lesson 1 - Introduction to Genetics.pptx
GenBio2 - Lesson 1 - Introduction to Genetics.pptxBerniceCayabyab1
 
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfBehavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfSELF-EXPLANATORY
 

Recently uploaded (20)

Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
 
BREEDING FOR RESISTANCE TO BIOTIC STRESS.pptx
BREEDING FOR RESISTANCE TO BIOTIC STRESS.pptxBREEDING FOR RESISTANCE TO BIOTIC STRESS.pptx
BREEDING FOR RESISTANCE TO BIOTIC STRESS.pptx
 
Vision and reflection on Mining Software Repositories research in 2024
Vision and reflection on Mining Software Repositories research in 2024Vision and reflection on Mining Software Repositories research in 2024
Vision and reflection on Mining Software Repositories research in 2024
 
Call Girls In Nihal Vihar Delhi ❤️8860477959 Looking Escorts In 24/7 Delhi NCR
Call Girls In Nihal Vihar Delhi ❤️8860477959 Looking Escorts In 24/7 Delhi NCRCall Girls In Nihal Vihar Delhi ❤️8860477959 Looking Escorts In 24/7 Delhi NCR
Call Girls In Nihal Vihar Delhi ❤️8860477959 Looking Escorts In 24/7 Delhi NCR
 
Volatile Oils Pharmacognosy And Phytochemistry -I
Volatile Oils Pharmacognosy And Phytochemistry -IVolatile Oils Pharmacognosy And Phytochemistry -I
Volatile Oils Pharmacognosy And Phytochemistry -I
 
Scheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxScheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docx
 
Transposable elements in prokaryotes.ppt
Transposable elements in prokaryotes.pptTransposable elements in prokaryotes.ppt
Transposable elements in prokaryotes.ppt
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
 
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxTHE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
 
OECD bibliometric indicators: Selected highlights, April 2024
OECD bibliometric indicators: Selected highlights, April 2024OECD bibliometric indicators: Selected highlights, April 2024
OECD bibliometric indicators: Selected highlights, April 2024
 
BIOETHICS IN RECOMBINANT DNA TECHNOLOGY.
BIOETHICS IN RECOMBINANT DNA TECHNOLOGY.BIOETHICS IN RECOMBINANT DNA TECHNOLOGY.
BIOETHICS IN RECOMBINANT DNA TECHNOLOGY.
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real time
 
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
 
(9818099198) Call Girls In Noida Sector 14 (NOIDA ESCORTS)
(9818099198) Call Girls In Noida Sector 14 (NOIDA ESCORTS)(9818099198) Call Girls In Noida Sector 14 (NOIDA ESCORTS)
(9818099198) Call Girls In Noida Sector 14 (NOIDA ESCORTS)
 
Evidences of Evolution General Biology 2
Evidences of Evolution General Biology 2Evidences of Evolution General Biology 2
Evidences of Evolution General Biology 2
 
Call Us ≽ 9953322196 ≼ Call Girls In Lajpat Nagar (Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Lajpat Nagar (Delhi) |Call Us ≽ 9953322196 ≼ Call Girls In Lajpat Nagar (Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Lajpat Nagar (Delhi) |
 
Artificial Intelligence In Microbiology by Dr. Prince C P
Artificial Intelligence In Microbiology by Dr. Prince C PArtificial Intelligence In Microbiology by Dr. Prince C P
Artificial Intelligence In Microbiology by Dr. Prince C P
 
Neurodevelopmental disorders according to the dsm 5 tr
Neurodevelopmental disorders according to the dsm 5 trNeurodevelopmental disorders according to the dsm 5 tr
Neurodevelopmental disorders according to the dsm 5 tr
 
GenBio2 - Lesson 1 - Introduction to Genetics.pptx
GenBio2 - Lesson 1 - Introduction to Genetics.pptxGenBio2 - Lesson 1 - Introduction to Genetics.pptx
GenBio2 - Lesson 1 - Introduction to Genetics.pptx
 
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfBehavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
 

Dr. Hager Scholarship at Harvard (2012)

  • 1. DIAPH3 Governs the Cellular Transition to the Amoeboid Tumour Phenotype The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters. Citation Hager, Martin H., Samantha Morley, Diane R. Bielenberg, Sizhen Gao, Matteo Morello, Ilona N. Holcomb, Wennuan Liu, et al. 2012. Diaph3 governs the cellular transition to the amoeboid tumour phenotype. EMBO Molecular Medicine 4(8): 743-760. Published Version doi:10.1002/emmm.201200242 Accessed October 8, 2013 10:44:17 PM EDT Citable Link http://nrs.harvard.edu/urn-3:HUL.InstRepos:10589807 Terms of Use This article was downloaded from Harvard University's DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http://nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA (Article begins on next page)
  • 2. DIAPH3 governs the cellular transition to the amoeboid tumour phenotype Martin H. Hager1,2y,z, Samantha Morley1,2z, Diane R. Bielenberg2,3, Sizhen Gao4, Matteo Morello1,2,5, Ilona N. Holcomb6, Wennuan Liu7, Ghassan Mouneimne4, Francesca Demichelis8,9, Jayoung Kim1,2,5, Keith R. Solomon1,10, Rosalyn M. Adam1,2, William B. Isaacs11, Henry N. Higgs12, Robert L. Vessella13, Dolores Di Vizio1,2,5, Michael R. Freeman1,2,5,14* Keywords: cytoskeleton; EGFR; endocytosis; mesenchymal-to-amoeboid transition; metastasis DOI 10.1002/emmm.201200242 Received November 23, 2011 Revised March 19, 2012 Accepted March 28, 2012 Therapies for most malignancies are generally ineffective once metastasis occurs. While tumour cells migrate through tissues using diverse strategies, the signalling networks controlling such behaviours in human tumours are poorly understood. Here we define a role for the Diaphanous-related formin-3 (DIAPH3) as a non-canonical regulator of metastasis that restrains conversion to amoeboid cell behaviour in multiple cancer types. The DIAPH3 locus is close to RB1, within a narrow consensus region of deletion on chromosome 13q in prostate, breast and hepatocellular carcinomas. DIAPH3 silencing in human carcinoma cells destabi-lized microtubules and induced defective endocytic trafficking, endosomal accumulation of EGFR, and hyperactivation of EGFR/MEK/ERK signalling. Silencing also evoked amoeboid properties, increased invasion and promoted metastasis in mice. In human tumours, DIAPH3 down-regulation was associated with aggressive or metastatic disease. DIAPH3-silenced cells were sensitive to MEK inhibition, but showed reduced sensitivity to EGFR inhibition. These findings have implications for understanding mechanisms of metastasis, and suggest that identifying patients with chromosomal deletions at DIAPH3 may have prognostic value. INTRODUCTION Improving clinical options for metastatic disease requires an understanding of the molecular basis for the complex behaviours of metastatic cells. To date, however, few genetic lesions in human tumours that promote selection of metastatic variants have been identified. Tumour cell migration can lead to metastatic dissemination. The ability of carcinoma cells to transition from an adherent, epithelial phenotype to a migratory, mesenchymal phenotype (the epithelial-to-mesenchymal transition, EMT) is well estab-lished, as are many signalling networks responsible for this transformation (Kalluri & Weinberg, 2009). However, it has recently been recognized that cancer cells can further transition Research Article DIAPH3 and the amoeboid phenotype (1) Urological Diseases Research Center, Children’s Hospital Boston, Boston, MA, USA (2) Department of Surgery, Harvard Medical School, Boston, MA, USA (3) Vascular Biology Program, Children’s Hospital Boston, Boston, MA, USA (4) Department of Cell Biology, Harvard Medical School, Boston, MA, USA (5) Division of Cancer Biology and Therapeutics, Samuel Oschin Compre-hensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA (6) Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA (7) Center for Cancer Genomics, Wake Forest University, Winston-Salem, NC, USA (8) Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA (9) Centre for Integrative Biology, University of Trento, Trento, Italy (10) Department of Orthopaedic Surgery, Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA (11) Department of Urology and Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA (12) Department of Biochemistry, Dartmouth Medical School, Hanover, NH, USA (13) Department of Urology, University of Washington, Seattle, WA, USA (14) Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA *Corresponding author: Tel: þ1 310 423 7069; Fax: þ1 310 423 0139; E-mail: michael.freeman@cshs.org y Present address: R&D Division, Oncology Research Laboratories, Daiichi Sankyo Europe, Munich, Germany zThese authors contributed equally to this work. www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 743
  • 3. to an ‘amoeboid’ phenotype (called the mesenchymal-to-amoeboid transition, MAT), characterized by a rounded morphology, extensive actomyosin contractility, rapid exten-sion and retraction of membrane protrusions (blebs), and reduced dependence on proteolysis for migration (Friedl Wolf, 2010). Amoeboid cells are behaviourally plastic, move rapidly through extracellular matrices by deforming their shape, and express surface proteins more symmetrically than mesenchymal cells, thus facilitating opportunistic responses to changes in the microenvironment (Schmidt Friedl, 2010). While such characteristics enable cells to readily migrate through extra-cellular matrix (ECM), the clinical impact of the amoeboid phenotype is unknown. Additionally, EMT (Kalluri Weinberg, 2009) and MAT (Wolf et al, 2003) are reversible processes, and the ability of tumour cells to switch between these varied states presents challenges in defining critical signalling nodes where metastasis might be inhibited. While the amoeboid phenotype is potentially relevant to aggressive cancer (Sanz-Moreno Marshall, 2010), it has not been well studied in human tumours. Chromosome 13 is frequently altered in cancer, and the q-arm suffers predominantly from deletions (Beroukhim et al, 2010). The RB1 locus at 13q14.2 is thought to be the dominant tumour suppressor affected by copy number alterations. However, reports speculate that another tumour suppressor resides in this region, whose loss is associated with breast (Kainu et al, 2000) and invasive prostate (Dong et al, 2000) cancers, and whose restoration can inhibit metastatic dissemination (Hosoki et al, 2002). Proximal to RB1, at 13q21.2, is the DIAPH3 locus encoding the protein Diaphanous-related formin-3 [DIAPH3, (Peng et al, 2003)]. This protein belongs to a family of formin orthologs (Chalkia et al, 2008) that nucleate, elongate and bundle linear actin filaments and regulate microtubule dynamics (Bartolini Gundersen, 2010; Gaillard et al, 2011; Goode Eck, 2007). Formins are required for such fundamental processes as endocytic trafficking, mitosis, cell polarity and migration (Goode Eck, 2007). Despite the deregulation of these events in pathological states such as cancer, the role of DIAPH3 in disease is unexplored.We recently reported that genomic loss at DIAPH3 was linked to metastatic prostate cancer (Di Vizio et al, 2009). However, given the propensity for chromosomal loss along 13q in multiple cancers (Beroukhim et al, 2010), whether DIAPH3 deletions are driver or passenger lesions remained unresolved. We now demonstrate that genomic loss at DIAPH3 is associated with multiple tumour types, and that DIAPH3 resides at an important signalling node that controls the amoeboid phenotype. We show that DIAPH3 silencing disrupts micro-tubules, impairs endocytosis and prevents EGFR downregulation, thereby enhancing EGFR activity and that of its downstream effectors MEK and ERK. DIAPH3 deficiency also promotes motility, invasion and experimental metastasis and significantly correlates with aggressive disease in human tumours. RESULTS Loss of DIAPH3 with tumour progression To test whether genomic loss at DIAPH3 is a driver or passenger effect consequent to the general instability of chromosome 13q, we employed the genomic identification of significant targets in cancer (GISTIC) platform (Beroukhim et al, 2010). GISTIC identified regions of somatic copy number alterations (SCNA) in a pooled analysis of 26 different cancer types, most of which are not associated with previously validated tumour suppressor genes. We hypothesized that significant (q-value 0.25) focal SCNA harbouring a tumour suppressor would emerge as a consensus area across these cancers. Among the deletion peaks on chromosome 13, one containing three genes, protocadherin 17 (PCDH17), tudor domain-containing protein 3 (TDRD3) and DIAPH3, was located in a region common to three carcinomas (prostate, breast and hepatocellular, Fig 1A). DIAPH3, near the centre of this consensus region, was deleted in 32% of prostate, 46% of breast and 31% of hepatocellular carcinomas (Support-ing Information Fig S1A). Together with the associated q-value range of 0.052q0.132, these data suggest that DIAPH3 deletions are enriched by selective pressure. In support of this hypothesis, Oncomine expression profiles indicated DIAPH3 mRNA levels were lower in high stage prostate cancer (PCa) and in hepatocellular and invasive breast carcinomas than in normal tissue [Fig 1B–D; (Finak et al, 2008; Li et al, 2006; Mas et al, 2009)]. Given the prevalence of DIAPH3 lesions in multiple tumour types, we examined DIAPH3 status in metastases, using PCa as a representative disease model. Metastatic PCa lesions can originate from a single cell (Liu et al, 2009) and may lie dormant for many years (Wikman et al, 2008). For these reasons, we characterized the DIAPH3 status of disseminated tumour cells (DTCs) isolated from bone marrow of PCa patients (Holcomb et al, 2008). Deletions were more frequent in DTCs with either prostate-confined (31%) or metastatic (46%) disease Research Article DIAPH3 and the amoeboid phenotype Figure 1. Genomic loss at DIAPH3 in invasive and metastatic tumours. A. The DIAPH3 locus on chromosome 13q21.2 is located in a focal peak region of significant deletion (q0.25) common in prostate, hepatocellular and breast carcinomas. Only three genes, PCDH17, TDRD3 and DIAPH3, are focally deleted in all cancer subtypes. B-D. DIAPH3 mRNA levels in prostate (PCa, stage T2/T3), hepatocellular and invasive breast carcinomas compared to normal tissue. E. Frequency of loss at DIAPH3 (grey line) in DTCs aspirated from bone marrow of patients with prostate-confined cancer (n¼48, blue) and advanced PCa (n¼11, red). Determined by array CGH and compared to copy number status in the primary tumours (n¼9, green). The chromosome band for each genomic position is shown; tick marks above indicate gene locations on chromosome 13. F. DIAPH3 copy number status grouped by Gleason score and compared with metastases. G. DIAPH3 copy number analysis in matched primary tumours and metastases (Met) reveals significant loss in metastatic disease. H. DIAPH3 gene copy number in PCa samples grouped by Gleason Score in comparison to metastases (Met). 744 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
  • 4. Research Article Martin H. Hager et al. B C Hepatocellular carcinoma D Breast carcinoma n=6 n=53 N Normalized expression units DIAPH3 0.5 -0.5 -1.5 invasive BCa Normalized expression units DIAPH3 -1.8 -2.0 -2.2 -2.4 n=19 n=38 N HCa Prostate carcinoma Normalized expression -1.2 units DIAPH3 n=4 n=4 n=12 T2 -1.5 -1.8 -2.1 N T3 PCa A n=53 n=102 n=12 n=12 n=36 Advanced Disease Prostate-confined Disease -0.5 Met Gleason Score 0.2 0.1 0 -0.1 -0.2 -0.3 -0.4 DIAPH3 log2 copy number units -0.5 6 7 8 9 0.1 20 10 F G DIAPH3 log2 copy number units 0 -0.1 -0.2 -0.3 -0.4 n=181 n=37 Primary Met Site 80 70 60 50 40 30 20 10 % of patients with deletion at DIAPH3 locus Grouped by Gleason Score n = 24 n = 63 n = 43 6 7 8 Gleason Score Metastasis 0 n = 18 Mbp H 50 40 30 RB1 DIAPH3 0 50 55 60 65 70 75 % of samples Primary Tumour Disseminated Tumour Cells E Figure 1. www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 745
  • 5. than in primary tumours (10%, Fig 1E), suggesting that DIAPH3 loss is related to dissemination from the primary site. To examine this possibility, we evaluated DIAPH3 copy number in primary tumours and metastases of 148 prostate cancers by Affymetrix Genome-Wide Human SNP Array 6.0 (Liu et al, 2009). Loss at DIAPH3 was increasingly prevalent with progression, occurring in 25% of low grade (Gleason score 6), 33% of intermediate grade (Gleason score 7), 40% of high grade (Gleason score 8), and 61% of metastatic tumours (Fig 1F, Supporting Information Fig S1B). Oncomine copy number data confirmed DIAPH3 genomic loss and its significant correlation with disease progression, which increased from primary tumour to metastatic lesions and with increasing Gleason score [Fig 1G and H; (Taylor et al, 2010)]. DIAPH3 knockdown induces an amoeboid phenotype in transformed cells In order to examine the biological consequences of DIAPH3 loss, we employed RNAi against DIAPH3 with multiple independent shRNAs or siRNAs (Supporting Information Fig S2A, Fig 6C), which efficiently reduced DIAPH3 protein levels but did not affect the related proteins DIAPH1 and DIAPH2 (Supporting Information Fig S2B). Because DIAPH3 loss was observed in breast cancers (Fig 1A and D), we employed the disease-relevant human mammary epithelial cell (HMEC) model. While stable DIAPH3 knockdown did not alter HMEC cell morphology in 2D culture, it modestly enhanced proliferation (Fig 2A, Supporting Information Fig S2C and D). In reconstituted basement membrane gels (3D culture), DIAPH3 silencing induced enlarged acinar structures with aberrant architecture and distribution of Laminin V (Fig 2A, Supporting Information Fig S3A and B), resembling that seen with ErbB2 activation (Debnath et al, 2002). These results suggest that in normal epithelia, DIAPH3 may act to suppress oncogenic behaviour. Next, we investigated the consequences of DIAPH3 loss in the context of HRAS-induced cellular transformation. We generated stable populations of HRASV12-transformed HMECs in which DIAPH3 was silenced (Supporting Information Fig S3C). In 2D culture, HMEC-HRASV12 cells appeared spindle-shaped, con-sistent with EMT (Fig 2B, top). In contrast, DIAPH3-depleted HMEC-HRASV12 cells were refractile, with rounded morpholo-gies (Fig 2B). Time-lapse video microscopy demonstrated that these cells displayed abundant, reversible eruption of blebs, reduced cell–cell contact, and increased rates of random motility, in comparison to HMEC-HRASV12 cells (Fig 2C and D, Supporting Information Movies S1, S2). These features are indicative of amoeboid behaviour (Wolf et al, 2003). In 3D culture, HMEC-HRASV12 produced large cell aggregates (acini) with penetration of the matrix by cords of spindle-shaped cells (Fig 2B, top inset, Supporting Information Fig S3D). In contrast, DIAPH3-deficient HRASV12 cells produced acini that progressively and dramatically lost structural integrity, coin-ciding with extensive migration of round cells into the matrix (Fig 2B, lower insets, Supporting Information Fig S3D). Significantly, while HRASV12-HMEC cells were poorly invasive through collagen I, DIAPH3 knockdown in this cell background promoted invasion (Fig 2E). These findings indicate that, when combined with activated HRAS, DIAPH3 down-regulation promotes an amoeboid phenotype and increases invasion. To determine whether this amoeboid transition can be similarly induced in genuine cancer cells, we silenced DIAPH3 in DU145 PCa cells. As in the genetically defined HRASV12- HMEC model, DIAPH3 knockdown in DU145 cells induced a rounded morphology and formation of membrane blebs when cells were embedded in collagen (Fig 2F). In line with this promotion of amoeboid behaviour (Fig 2G), DIAPH3 knock-down enhanced the invasiveness of DU145 cells (Fig 2H, Supporting Information Fig S3E). DIAPH3-deficient cells were hypersensitive to chemo-attractants, including EGF. Amoeboid cells display weakened substrate adhesions, allowing high cell velocities (Schmidt Friedl, 2010). This fast ‘gliding’ mode can be driven by Rho-kinase (ROCK)-dependent force generation through myosin light chain (MLC2) phosphor-ylation, and is distinct from integrin-mediated traction force generation characteristic of mesenchymal motility (Friedl Wolf, 2010). DIAPH3 silencing promoted focal adhesion disassembly, as evidenced by an altered pattern of focal adhesion kinase (FAK) phosphorylation [Fig 3A, 3B (Hamadi et al, 2005)], and also evoked high levels of phosphorylated MYPT1, a ROCK substrate (Fig 3C and D), and MLC2 (Fig 3E). In DIAPH3-deficient cells, both activated and total MLC2 were enriched at the cell cortex (Fig 3F, Supporting Information Fig S4A–C), where cortical MLC2 colocalized with phalloidin. In unsilenced cells, cortical MLC2 was less pronounced and co-localized insteadwith phalloidin in stress fibres. These findings are consistent with the ROCK-mediated cortical localization of MLC2 reported to be displayed by amoeboid cells (Pinner Sahai, 2008; Sahai Marshall, 2003; Wyckoff et al, 2006). Research Article DIAPH3 and the amoeboid phenotype Figure 2. DIAPH3 knockdown induces an amoeboid phenotype in transformed cells. A. Morphology of DIAPH3-depleted HMEC, compared to control cells. Plastic (2D), basement membrane cultures (3D). Scale¼200 mm. B. HRASV12 cells grown in 2D adopt a round, refractile appearance when DIAPH3 is silenced. Scale¼100 mm. HMEC-HRASV12 cells form cell aggregates with cord-like protrusions in 3D. DIAPH3 depletion causes junctional instability, with migration of single cells into the surrounding matrix (inset, right). Scale¼200 mm. C. Representative trajectories of control (green) and DIAPH3-deficient (red) HMEC-HRASV12 cells. D. Quantification of migration speed, determined from C. E. DIAPH3-silenced HRASV12-HMEC display increased invasiveness. F. Growth of DIAPH3-deficient and control DU145 cells on a thick layer of collagen I. Arrowheads mark cell surface blebbing (top). Visualization of the cytoskeleton with phalloidin by immunofluorescence (IF) shows prominent cortical actin in DIAPH3-deficient cells (arrowhead, bottom). Scale¼50 mm. G. Increase in amoeboid morphology following expression of DIAPH3 siRNA. H. Invasion of collagen I by DU145 cells, with FBS or EGF as chemo-attractants, is increased in DIAPH3-deficient cells (N2 independent trials). See also Supporting Information Movies S1 and S2. 746 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
  • 6. Research Article Martin H. Hager et al. B HMEC HMEC-RasV12 2D 3D 2D 3D 3D (inset) C D Migration E Distance (μm) Distance (μm) Control shRNA DIAPH3 shRNA Fluorescence (RFU X 103) Invasion 50 40 30 20 10 0 Control shRNA HMEC HRASV12 Control shRNA DIAPH3 shRNA 4 3 2 1 0 Speed (μm/min) HMEC HRASV12 DU145 Invasion G Morphology F H Control shRNA DIAPH3 shRNA Control shRNA DIAPH3 shRNA FBS 6h FBS 12h EGF 12h Fluorescence (RFU X 103) 80 60 40 20 0 DIAPH3 shRNA A F-actin F-actin Control shRNA DIAPH3 shRNA1 DIAPH3 shRNA2 Control shRNA DIAPH3 shRNA1 DIAPH3 shRNA2 Control shRNA DIAPH3 shRNA1 DIAPH3 shRNA2 Control shRNA DIAPH3 shRNA1 DIAPH3 shRNA2 Control shRNA DIAPH3 shRNA1 DIAPH3 shRNA2 100 80 60 40 20 0 amoeboid cells/field (percent of total) Control siRNA DIAPH3 siRNA DU145 DU145 Figure 2. www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 747
  • 7. A B Control shRNA MYPT1 DIAPH3 regulates EGFR trafficking and microtubule stability We next sought to examine the mechanisms underlying induction of the amoeboid transition by DIAPH3 knockdown. Formins localize to endosomes and regulate vesicular trafficking (Fernandez-Borja et al, 2005; Gasman et al, 2003; Wallar et al, 2007). Because endocytosis governs the spatiotemporal regula-tion of receptor tyrosine kinases (RTKs), and chemotactic sensitivity to EGF was enhanced in DIAPH3-silenced cells (Fig 2H), we hypothesized that DIAPH3 may regulate turnover and trafficking of EGFR. Enforced expression of DIAPH3 markedly accelerated the kinetics of EGFR turnover and receptor Control siRNA Control shRNA DIAPH3 shRNA 12 10 8 6 4 2 20 16 12 8 4 pMLC2 (S19) Phalloidin inactivation in response to EGF and under steady-state conditions (Fig 4A, 4B). Further supporting a functional relationship, DIAPH3 and EGFR co-localized in endosomes (unpublished observations). Thus, we examined the influence of DIAPH3 on endocytic trafficking of EGFR. In the absence of EGF and the presence of endogenous DIAPH3, EGFR co-localized at the plasma membrane with the recycling marker Rab11 (Fig 4C). In response to ligand, EGFR translocated to a peri-nuclear region and co-localized with the early endosome marker Rab5 (Fig 4D). In contrast, DIAPH3 silencing promoted association of EGFR with endosomes enriched in Rab11, Rab5 Research Article DIAPH3 and the amoeboid phenotype Focal adhesions per cell DIAPH3 siRNA pMLC2 MLC2 DIAPH3 -Actin Control shRNA DIAPH3 shRNA F DU145-Control shRNA DU145-DIAPH3 shRNA pMLC2 (S19) Phalloidin Dapi Merge Dapi Merge C ROCK DIAPH3 siRNA Control siRNA pMYPT1 MYPT1 pMYPT1 pMYPT1/MYPT1 (fold of control) 0 ROCK DIAPH3 shRNA D 0 p0.0001 Control siRNA pFAK (Y397) DIAPH3 siRNA pFAK (Y397) E Figure 3. DIAPH3 knockdown promotes biochemical features of the amoeboid transition. A. The distribution of focal adhesions in COS7 cells, detected by IF with a phospho-FAK(Y397) antibody, is reduced in DIAPH3-deficient cells. Scale¼20 mm. B. Quantitation of focal adhesions in A. C-E. Enhanced phosphorylation of MYPT1 (C,D) and MLC2 (E) in DIAPH3-deficient DU145 cells indicates high ROCK activity. F. Distribution of active (phosphorylated) MLC2 and F-actin (phalloidin) in DU145 stable lines. Scale¼10mm (N2 independent trials). 748 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
  • 8. and EEA1 (Fig 4C–E), even in absence of ligand. These findings suggest that loss of DIAPH3 disrupts vesicular transport, leading to EGFR accumulation in endosomes. To investigate how DIAPH3 affects endosomal trafficking of EGFR, we assessed its influence on the actin and micro-tubule (MT) cytoskeletons, which govern short- and long-range vesicular transport, respectively (Soldati Schliwa, 2006). While rearrangement of the actin cytoskeleton was associated with DIAPH3 silencing (Fig 3F, Supporting Information Fig S4C), no conspicuous actin defects were detected. Combined with the observation that endosomes did not accumulate at the cell cortex but were dispersed throughout the cytoplasm in DIAPH3- deficient cells (Fig 4C–E), these data suggest that DIAPH3 loss does not significantly influence short-range EGFR trafficking. In addition to regulating actin dynamics, formins also promote MT stability (Bartolini et al, 2008). Enforced DIAPH3 markedly increased tubulin acetylation (Fig 5A, Supporting Information Fig S5A), an indication of MT stabilization (Verhey Gaertig, 2007). Conversely, DIAPH3 silencing decreased tubulin acetylation and was associated with MT fragmentation (Fig 5B–G, Supporting Information Fig S5B). Notably, treatment of cells with the MT depolymerizing agent nocodazole phenocopied the accumulation of EGFR in internal vesicles and some amoeboid features (Supporting Information Fig S5C and D) induced by DIAPH3 knockdown. These findings imply that DIAPH3 deficiency disrupts MT stability and endosome processing, thereby sequestering and preserving EGFR in early endosomes. Disrupted trafficking coincided with EGFR hyper-activation, as evidenced by increased auto-phosphorylation of activating residues and decreased phosphorylation of the inhibitory Thr669 site (Fig 6A). EGFR activation was enhanced in DIAPH3-silenced cells in response to EGF (Supporting Information Fig S6A). DIAPH3 deficiency also increased phosphorylation of the EGFR effectors MEK and ERK (Fig 6A and B, Supporting Information Fig S6A), which inversely and dose-dependently correlated with DIAPH3 levels (Fig 6C, Supporting Information Fig S6B). Even under serum-free conditions, DIAPH3 depletion increased ERK phosphorylation sixfold (Fig 6D), and potentiated the response to EGF (Fig 6E, Supporting Information Fig S6C), with a twofold to fourfold increase in phosphorylated ERK at each time point post-stimulation. This ligand-independent hyperactivation of EGFR and MEK/ERK is consistent with reports linking early endosome-localized EGFR to sustained activation of the RAF/ MEK/ERK pathway (Sorkin von Zastrow, 2009). Collectively, Research Article Martin H. Hager et al. these findings support an important role for DIAPH3 in regulation of EGFR trafficking and signalling. Sensitivity of DIAPH3-deficient cells to anti-neoplastic agents Because DIAPH3 silencing induces amoeboid behaviour and strongly activates ERK, we tested the relevance of MEK/ERK signalling in this scenario. Treatment of DIAPH3-deficient DU145, HMEC-HRASV12 and A375P melanoma cells with the MEK1/2 inhibitor PD98059 converted cells to a more mesench-ymal morphology (Fig 7A, Supporting Information Fig S7A). Pharmacologic reversion of amoeboid features could be reversed by drug washout (unpublished observations). A375P has been used as a model to study amoeboid properties (Sanz- Moreno et al, 2008). Collectively, these data indicate that MEK/ ERK pathway activation can contribute to amoeboid behaviour in diverse cell backgrounds. EGFR is a frequent target for therapeutic intervention. However, responsiveness to tyrosine kinase inhibitors (TKI, e.g. gefitinib)may be dictated by the receptor’s subcellular localization (Huang et al, 2009). Because DIAPH3 knockdown modulates EGFR localization and activity, we asked whether DIAPH3 deficiency affects TKI sensitivity. In control DU145 cells, EGF stimulation induced amoeboid features (blebbing), which was suppressed by gefitinib (Fig 7B and C, Supporting Information Movie S3). In contrast, the amoeboid phenotype was constitutive and ligand-independent in DIAPH3-silenced cells. Notably, these cells were unresponsive to the TKI. These findings suggest that DIAPH3 loss alters sensitivity to EGFR inhibitors. Taxanes such as paclitaxel and docetaxel are first-line chemotherapies for metastatic breast and prostate cancers, which we show are prone to DIAPH3 loss (Fig 1). However, development of resistance limits their efficacy in patients. Taxanes stabilize MTs, suggesting their potential to down-regulate EGFR similarly to DIAPH3. We observed, however, that EGFR activation persisted in the presence of docetaxel (Supporting Information Fig S7B) and paclitaxel (unpublished observations), in line with reports that these agents disrupt endosomal EGFR trafficking (Sonee et al, 1998). DIAPH3 suppresses the amoeboid phenotype Induction of the amoeboid phenotype by DIAPH3 knockdown suggests that enforced DIAPH3 expression may counteract this transition. We stably expressed GFP-DIAPH3 in PC3 PCa cells and in U87 glioblastoma cells. Of note, U87 expressed the lowest levels of DIAPH3 among the cell lines we tested. U87 and PC3 Figure 4. DIAPH3 regulates endocytic trafficking of EGFR. A. Enforced DIAPH3 accelerates EGFR turnover. COS7 cells expressing empty vector (Vo) or DIAPH3 were serum-depleted overnight, treated with EGF for the indicated times, and lysates blotted with the antibodies shown. B. GFP-DIAPH3-positive COS7 cells show strongly reduced EGFR levels (lower panel, arrowhead) in comparison with GFP-transfected control cells (upper panel) or adjacent untransfected cells. Scale¼20 mm. C. Left, Co-localization of Cy3-labeled EGFR with FITC-labelled Rab11 in DU145 cells expressing DIAPH3 siRNAs. Right, Cell peripheries are shown in grey, and areas of co-localization in black. Insets show predominance of EGFR at the plasma membrane (top) or in endosomes (bottom). Scale¼20mm. D. Left, Co-localization of Cy3-labeled EGFR with FITC-labelled Rab5 upon ligand binding (þEGF). DIAPH3 loss evokes an increase in EGFR internalization in the absence of ligand (EGF), as shown by accumulation in endosomes (insets and arrowheads) and co-localization with Rab5. Scale¼20 mm. Right, Quantitation of EGFR-enriched endosomes. E. Cy3-labeled EGFR, which is localized at the plasma membrane in DU145 cells expressing control siRNA, is internalized and co-localizes with the FITC-labelled early endosome marker EEA1 in DU145 cells expressing DIAPH3 siRNAs. Scale¼10mm (N2 independent trials). www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 749
  • 9. Research Article DIAPH3 and the amoeboid phenotype EGF: pEGFR (Y1086) EGFR DIAPH3 -actin Vo DIAPH3 A B C E Control siRNA DIAPH3 siRNA 180 150 120 90 60 30 0 Control siRNA - EGF + EGF EGFR-positive endosomes per cell DIAPH3 siRNA EGFR Rab11 Control siRNA EGFR EGFR Rab5 Rab5 Merge Merge DIAPH3 siRNA EGFR Rab5 Merge - EGF + EGF - EGF + EGF EGFR EEA1 EGFR EEA1 Common pixels Control shRNA DIAPH3 shRNA EGFR Rab11 EGFR/GFP-DIAPH3 GFP-DIAPH3 GFP EGFR EGFR/GFP EGFR D EGFR Rab5 Merge Figure 4. 750 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
  • 10. Research Article Martin H. Hager et al. Control shRNA Ac-tubulin DIAPH3 shRNA DIAPH3 -actin A Control shRNA DIAPH3 shRNA p = 0.0011 Ac-tubulin / -actin (fold of control) 1.0 0.5 0 Ac-tubulin DIAPH3 -actin Vo DIAPH3 p = 0.038 -tubulin Ac-tubulin / -actin (fold of control) Vo DIAPH3 8 6 4 2 0 D Ac-tubulin fluorescence Control shRNA Tubulin pixels (rendered) DIAPH3 shRNA x y x y Pixel intensity, y plane Control shRNA Distance (pixels), x plane 12 9 6 0 100 200 300 400 500 3 DIAPH3 shRNA C B F Control shRNA DIAPH3 shRNA G Phalloidin Ac-tub Phalloidin Ac-tub Control shRNA DIAPH3 shRNA p = 0.0033 n = 488 Ac-tubulin Intensity (fold of control) 1.0 0.5 0 n = 577 Control shRNA DIAPH3 shRNA CTxB Ac-tub CTxB Ac-tub E Figure 5. DIAPH3 regulates microtubule topology and stability. A. Increased acetylated tubulin (Ac-tubulin) in PC3 cells stably expressing DIAPH3. B. Reduced Ac-tubulin levels in DU145 cells silenced for DIAPH3. C. Left, Ac-tubulin IF showing MT fragmentation in DIAPH3-deficient DU145 cells grown in 3D. Right, MT topology was rendered by ImageJ. Insets, cells stained with cholera toxin B (CTxB, green). Scale¼10 mm. D. Pixel intensity of C was quantified as a 2D contour plot, as a function of intensity in the x versus y planes (illustrated schematically, top). E. The pixel intensity of Ac-tubulin was quantified in silenced or unsilenced DU145 cells. Cell peripheries were outlined as in C, tubulin intensity integrated within the enclosed area, and average intensities in each condition determined. Average intensity values determined from three independent trials (Student’s t-test, p¼0.0033). N¼total cell number. F,G. DU145 cells expressing control or DIAPH3 shRNAs were stained with Ac-tubulin following staining with rhodamine-phalloidin (F) or FITC-CTxB (G), and imaged by fluorescence microscopy. Scale¼10mm (N2 independent trials). www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 751
  • 11. pEGFR (T669) pEGFR (Y992) pERK1/2 1 0.3 1 1.6 Mock ERK1/2 8 pMEK1/2 (S217/221) MEK1/2 DIAPH3 -actin ERK1/2 both were phenotypically amoeboid, with numerous membrane blebs and rounded morphologies (Fig 8A and B, Supporting Information Fig S8A). Enforced DIAPH3 altered the phenotype of both cell lines, suppressing amoeboid blebbing and inducing formation of prominent lamellipodia, a typical mesenchymal feature (Fig 8A, B and F, Supporting Information Fig S8A). DIAPH3 also increased levels of the mesenchymal marker N-cadherin (Fig 8C), in concert with increased stress fiber formation (Fig 8E, Supporting Information Fig S8B), suggesting that DIAPH3 promotes an amoeboid to mesenchymal transition. The data shown above position DIAPH3 within the EGFR pathway. Consistent with this, we identified an EGF-sensitive phospho-serine at position 624 in the DIAPH3 primary sequence by tandem mass spectrometry (Supporting Information Fig S9A). S624 is located within the last of five polyproline/SRC homology three binding motifs in the FH1 domain (Fig 8D). This site was validated by stable isotope labelling (Supporting Control siRNA Information Fig S9B and C) and confirmed to be EGF-responsive using a custom phosphosite-specific antibody (Supporting Information Fig S9D and E). DIAPH3 function was modulated by its phosphorylation status at S624. A phospho-null mutant at this site (S624A) promoted stress fibre formation, a mesench-ymal characteristic (Fig 8E), and suppressed amoeboid blebbing (Fig 8F) to a greater degree than the unmodified protein, while a phospho-mimetic mutant (S624E) was impaired in both activities (Fig 8E and F). These findings support the conclusion that DIAPH3 suppresses amoeboid blebbing and is functionally inhibited by phosphorylation at S624, which occurs in response to EGF treatment (Supporting Information Fig S9E). DIAPH3 loss promotes metastasis and is associated with metastatic human prostate cancer Our findings thus far suggest that DIAPH3 loss promotes motility, invasion, and increased oncogenic signalling, all pre-requisites for Research Article DIAPH3 and the amoeboid phenotype pEGFR (Y1068) EGFR Control siRNA DIAPH3 siRNA pEGFR (Y1086) ERK1/2 1 4.8 1 2.0 DIAPH3 -actin 1 4.2 DIAPH3 Control siRNA DIAPH3 siRNA1 DIAPH3 siRNA2 pERK1/2 C A D 10 pERK/ERK (fold of control) 6 4 2 0 DIAPH3 siRNA1 DIAPH3 siRNA2 B Control shRNA DIAPH3 shRNA DIAPH3 100 80 60 40 20 0 Relative DIAPH3 level (%) pERK1/2 ERK1/2 Control siRNA DIAPH3 siRNA #1 #2 #3 #4 E Control siRNA DIAPH3 siRNA 50 40 30 20 10 0 0 2 4 6 8 10 -tubulin DIAPH3 siRNA1 Control siRNA EGF (min): 0 2 5 10 0 2 5 10 pERK1/2 pERK signal intensity (a.u.) P=0.0068 Figure 6. DIAPH3 knockdown enhances EGFR signalling. A. EGFR is active in DIAPH3-silenced COS7 cells, as shown by increased tyrosine phosphorylation at activating sites and decreased phosphorylation of the T669 inhibitory site. B. Enhanced pMEK1/2 in DIAPH3-depleted DU145 cells. C. Targeting of DIAPH3 with four independent siR-NAs in COS7 leads to 90% depletion, which inversely correlates with pERK1/2 levels. D. DIAPH3 depletion in serum-depleted cells enhances pERK1/2 levels. E. Acute, sustained phosphorylation of ERK1/2 in response to EGF in DIAPH3-depleted versus control cells (two-way ANOVA, p¼0.0068; N2 independent trials). 752 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
  • 12. 80 60 40 HMEC HRASV12 A375P 100 80 60 40 20 100 80 60 40 20 0 DIAPH3 shRNA2 metastasis. Consistentwith this hypothesis, superficial pulmonary metastases in nudemice, induced by tail vein injection of DIAPH3- silenced DU145 cells, were enhanced relative to unsilenced cells (Fig 9A and B). Both the number and size of metastatic foci were potentiated by DIAPH3 knockdown (Fig 9B and data not shown). Additionally, large tumour thrombotic emboliwere observed only in lung sections from mice that underwent injection of DIAPH3- silenced cells (Fig 9C). Detection of human cytokeratin 18 (CK18) in the lung metastases demonstrated the lesions’ human origin (Fig 9D). This was further confirmed by puromycin resistance of cells from dissociated lesions in cell culture. Together, these findings indicate that DIAPH3 silencing enhances experimental metastasis in mice. Lastly, we examined expression of the DIAPH3 protein with PCa progression using a cohort of 90 human prostate specimens in a tissue microarray format and a validated anti-DIAPH3 antibody (Supporting Information Fig S2B). While DIAPH3 protein levels did not demonstrably change between benign and organ-confined carcinoma, we observed a dramatic reduction of DIAPH3 protein levels in metastatic lesions in comparison with normal tissue (p¼0.018) and organ-confined tumours (p¼0.007, Fig 9E and F). These results are in agreement with our genomic analyses (Fig 1E–H) and indicate that loss of the DIAPH3 locus functionally results in significant loss of the protein in human metastatic disease. 100 80 60 40 20 DISCUSSION Research Article Martin H. Hager et al. Control shRNA DIAPH3 shRNA1 This study provides evidence that the formin DIAPH3 belongs to a novel class of metastasis suppressors that inhibits conversion to an amoeboid phenotype. Inactivation of this gene appears relevant to several human malignancies, including prostate and breast carcinomas. We identified a consensus area of significant recurrent deletion on chromosome 13 encompassing the DIAPH3 locus and showed that DIAPH3 genomic loss and/or decreased DIAPH3 expression occurs in organ-confined tumours, but occurs at higher frequency in advanced disease, circulating prostate tumour cells, and metastatic lesions. We show that DIAPH3 silencing enhances tumour cell invasion, promotes amoeboid features in disparate cell backgrounds, and enhances experimental metastasis in vivo. This is the first example of a genomic lesion affecting a direct cytoskeletal regulator that governs the amoeboid transition. Because amoeboid behaviour enables cells to squeeze through gaps in the fibrillar matrix, amoeboid cells are proposed to possess a higher proclivity to disseminate and metastasize (Sanz-Moreno Marshall, 2010). We provide evidence that DIAPH3 deficiency promotes a wide range of amoeboid characteristics, and can cooperate with a canonical activated oncogene. In a mammary epithelial background, activated 20 DMSO PD98059 DMSO PD98059 DMSO PD98059 amoeboid cells / field (percent of total) 0 DU145 0 Control EGF EGF + Gefitinib amoeboid cells / field (percent of total) Control shRNA DIAPH3 shRNA B 0 Control EGF EGF + Gefitinib DIAPH3 shRNA Control shRNA C A Figure 7. Sensitivity of amoeboid features to MEK1/2 and EGFR inhibition. A. Amoeboid features induced by DIAPH3 silencing in human HMEC-HRASV12, A375P melanoma and DU145 cells can be reverted by PD98059 (50 mM). N2 independent trials. B,C. Amoeboid blebbing induced by EGF (10 nM) in control cells is sensitive to the EGFR inhibitor Gefitinib (2 mM), while induction by DIAPH3 silencing is less sensitive to both treatments, (C) Representative micrographs, (B) See also Supporting Information Movie S3. www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 753
  • 13. A PC3 B U87 DIAPH3 S p = 0.0012 200 150 100 50 HRAS alone induced EMT; in contrast, DIAPH3 silencing in the context of activated HRAS resulted in an amoeboid transition consisting of a dramatic morphologic transformation in base-ment membrane cultures and high invasive potential (Fig 2). These findings suggest that the oncogenic background of tissues in which DIAPH3 is lost makes an essential contribution to tumour behaviour. GFP GFP-DIAPH3 2.5 p = 0.0048 GFP p = 0.0017 p = 0.0032 2.0 1.5 1.0 S624 20 15 10 5 We also showed that an endosomal trafficking defect can elicit amoeboid behaviour. We propose a model (Fig 10) whereby DIAPH3 loss causes cytoskeletal disruption, inhibits endocytic down-regulation of RTKs, and leads to persistent activation of downstream effectors. That DIAPH3 down-regulation enhances endosomal accumulation of EGFR is notable given recent reports implicating endocytic trafficking Research Article DIAPH3 and the amoeboid phenotype E Stress fiber formation HeLa Degree of induction (percent of control) p 0.0001 p=0.0291 F Amoeboid blebbing U87 Membrane blebs per cell p 0.0001 GFP GFP-DIAPH3 C D DIAPH3: - + N-cadherin GFP-DIAPH3 GFP 0.5 -actin 0 N-cadherin / -actin (fold of control) GFP-DIAPH3 GBD DID DD CC FH1 FH2 DAD GFP-DIAPH3 variants GFP-DIAPH3 variants GFP GFP-DIAPH3 GFP-DIAPH3(S624A) GFP-DIAPH3(S624E) GFP GFP-DIAPH3 GFP-DIAPH3(S624A) GFP-DIAPH3(S624E) 0 0 Figure 8. DIAPH3 expression suppresses the amoeboid phenotype. A,B. Micrographs of PC3 (A) and U87 (B) cells expressing GFP or GFP-DIAPH3. Arrows indicate prevalence of amoeboid cells in PC3- or U87-GFP (left panels), and of mesenchymal (lamellopodia-enriched) cells in PC3- or U87-GFP-DIAPH3 (right panels). C. N-cadherin is significantly upregulated in cells expressing GFP-DIAPH3. D. Domain structure of DIAPH3 and location of the S624 phosphosite within the FH1 domain. E. Quantitation of stress fiber formation in HeLa cells expressing GFP or DIAPH3 mutants and stained with phalloidin. F. Quantitation of amoeboid blebbing in U87 cells expressing GFP or DIAPH3 mutants and stained with CTxB (N2 independent trials). 754 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
  • 14. Normal/Benign Carcinoma Metastasis 100 in tumour suppression (Mosesson et al, 2008) and demonstrat-ing increased signalling from EGFR, VEGFR and c-MET if endosome processing is compromised (Joffre et al, 2011; Lanahan et al, 2010; Wang et al, 2009). Our findings suggest that amoeboid behaviour is a disease-relevant outcome of the Research Article Martin H. Hager et al. C Tumour thrombus DIAPH3 shRNA HE HE 80 60 40 20 ability of DIAPH3-silenced tumour cells to usurp the endocytic machinery. Our model proposes that deregulated endosomal trafficking and signalling defects associated with DIAPH3 silencing arise from MT disruption. DIAPH3 loss induced MT instability and A Control shRNA DIAPH3 shRNA Control shRNA DIAPH3 shRNA Control shRNA CK18 CK18 D DIAPH3 shRNA Control shRNA DIAPH3 shRNA p = 0.0493 Metastases per lung B Normal/ Benign PCa Met % of samples High Low 0 E F Figure 9. Silencing of DIAPH3 promotes metastasis and is associated with metastatic disease in vivo. A. DIAPH3-silenced DU145 cells injected into the tail vein of nude mice produced large superficial pulmonary metastases (arrowhead). B. Quantification of lung metastases in mice injected with control or DIAPH3-silenced DU145 cells (Mann–Whitney U test). N¼10 mice/condition. C. Representative thromboembolus from lung sections from mice injected with DIAPH3-silenced DU145 cells. D. Representative lung sections of mice injected with cells expressing control or DIAPH3 shRNAs, stained with HE or an antibody to human CK18. Note presence of micrometastases in sections from DIAPH3 shRNA mice (arrowhead; N¼2 independent trials). E. DIAPH3 IHC staining of a human tissue microarray (TMA) containing cores with benign human prostate epithelium (Normal/Benign), prostate tumour tissue (Carcinoma) and tissue from metastatic lesions (Metastasis). High-power magnifications are shown (bottom). Scale¼200 mm. F. DIAPH3 expression is significantly decreased in metastases (Fisher’s exact test, p¼0.018/0.007). www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 755
  • 15. EGF EGFR Microtubules crotubule crotubul Amoeboid APH3 EGFR-enriched EGFR activation, in agreement with reports of a positive correlation between tubulin acetylation and EGFR degradation (Deribe et al, 2009; Gao et al, 2010). MT instability is emerging as a contributor to the amoeboid phenotype (Belletti et al, 2008; Berton et al, 2009). Our findings suggest that DIAPH3 is a key regulatory node in the transition between amoeboid and mesenchymal tumour cell phenotypes and that MT disruption may affect amoeboid and mesenchymal features at multiple levels. The sustained endosomal localization of EGFR consequent to DIAPH3 downregulation may be clinically relevant. Response of a tumour cell population to a TKI can be influenced by distribution of intracellular EGFR, such that some ‘sensitive’ cells display EGFR on the plasma membrane while ‘resistant’ cells exhibit perinuclear localization (Huang et al, 2009). Alternatively, while the ligand-bound receptor undergoes normal endocytic trafficking in ‘responsive’ cells, transport to lysosomes can be defective in ‘resistant’ cells (Nishimura et al, 2007). We observed similar responses with DIAPH3 deficiency, which markedly reduced sensitivity to a TKI. Our findings are suggestive that DIAPH3 loss can induce resistance to EGFR inhibitors. This possibility deserves further exploration. The RAS/MAPK axis is upregulated in 90% of metastatic PCa lesions (Taylor et al, 2010), and hyper-activation of ERK is implicated in PCa progression (Gioeli et al, 1999). However, it remains unclear how this pathway is activated, since compre-hensive profiling of prostate tumours did not reveal activating mutations in BRAF or HRAS (Burger et al, 2006; Thomas et al, 2007). It is notable, then, that in PCa cells DIAPH3 deficiency upregulates ERK activity. Our results raise the interesting possibility that MEK inhibitors may be useful to target advanced disease in patients with tumours with DIAPH3 loss. Two recent reports assessed DIAPH3 in the context of cell invasion. Lizarraga et al. demonstrated that DIAPH3 silencing inhibits formation of filopodia-like invadopodia, invasion and degradation of 3D-matrices (Lizarraga et al, 2009). We observed DIAPH3 loss to induce a switch to an amoeboid phenotype, in which dependence on proteases for invasion is reportedly reduced (Friedl Wolf, 2010). However, while DIAPH3 silencing suppressed invasion of MDA-MB-231 cells through Research Article DIAPH3 and the amoeboid phenotype Mesenchymal Lysosome / t tubulin heterodimer (disrupted MT) Early endosome DIAPH3: intact Focal adhesion maturation Stress fiber formation DIAPH3: lost Lack of focal adhesion maturation Cortical actomyosin contractility Membrane bleb formation Increased motility and invasion DIAPH3 Mic 1. 2. 3. 2. 1. 3. 4. 5. odimer disru endosome 1. A B Figure 10. Model of DIAPH3 perturbation and the amoeboid transition. A. When DIAPH3 is functional: (1) EGFR is interna-lized via endocytosis upon activation; (2) EGFR traffics in endosomes along microtubules to lysosomes or is recycled back to the plasma membrane; (3) ERK activity is attenuated following EGFR down-regulation. B. When DIAPH3 is lost or inactivated: (1) EGFR is internalized. (2) MT are destabilized, preventing both transport of EGFR from early endosomes to lysosomes, and receptor recycling. (3) Active EGFR accumulates in endosomes. (4) ERK activity is sustained. (5) Deregulation of proteins promoting amoeboid characteristics (e.g. MLC2, FAK) evokes cortical actomyosin contraction, focal adhesion turnover, and membrane blebbing. Broken arrows/lines indicate the presence of signalling intermediates. 756 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
  • 16. Matrigel (Lizarraga et al, 2009), we observed DIAPH3 silencing to promote invasion through collagen I. Using an siRNA screen for formin family regulators of membrane blebbing, Stastna et al. reported that DIAPH3 silencing inhibited bleb formation and promoted cell spreading in HeLa cells (Stastna et al, 2011). Of the multiple transcripts of the DIAPH3 locus, Isoform 1 mediated bleb formation, while an activated variant of Isoform 7 instead promoted filopodia. Consistent with the last observa-tion, we observed a significant reduction in filopodia following DIAPH3 silencing of HMEC-HRASV12 (unpublished observa-tions). We employed Isoform 7 for our studies, however DIAPH3 silencing potentiated bleb formation, reduced adhesion and increased rates of migration in COS7, DU145 and HMEC. Although genetic heterogeneity or different characteristics of diverse ECM used in these in vitro studies may play a role in these diverse effects, collectively they are consistent with the conclusion that DIAPH3 resides at an important signaling node that controls invasive behaviour. Importantly, the genomic loss data and other findings from human cohorts that we present here are consistent with the conclusion that DIAPH3 inactiva-tion is likely to promote aggressive behaviour in prostate, breast and possibly other tumour types. In the present study, we also identified a serine residue in the DIAPH3 FH1 domain that seems to result in inactivation of the protein when phosphorylated. This finding suggests that DIAPH3 might be inactivated by upstream signaling pathways in addition to gene disruption. Reports have speculated about the presence of a tumour suppressor at 13q21 that is independent of RB1. A recent study evaluated somatic homozygous deletions (HDs) at high resolution in 746 cancer cell lines (Bignell et al, 2010), Research Article Martin H. Hager et al. identifying an ‘unexplained’ HD cluster on chromosome 13 that exhibits a signature similar to known tumour suppressors. This cluster was separate from the dominating HD cluster affecting the RB1 locus and had not been assigned to a known tumour suppressor gene. DIAPH3 is a candidate non-canonical tumour suppressor in this region. In conclusion, identification of DIAPH3 as a protein capable of mediating the switch between mesenchymal and amoeboid phenotypes provides new insight into the molecular processes of metastasis, and may facilitate design of more effective strategies against advanced disease. MATERIALS AND METHODS Copy number analysis DNA copy number alterations were analysed with the Integrated Genomics Viewer using genome-wide GISTIC data (Beroukhim et al, 2007). DIAPH3 copy number status of PCa patients was analysed using comparative genomic hybridization (cCGH) and Affymetrix Genome- Wide SNP Array data (Liu et al, 2009). The frequency of DIAPH3 loss in circulating tumours cells was assessed using array CGH data (Holcomb et al, 2008). Immunohistochemistry and tissue microarrays The human prostate tissue microarray consisted of normal/benign (n¼16), prostate tumour (n¼22) and metastatic tissue cores (n ¼24). Immunohistochemistry was performed with DIAPH3 (HNH3.1) or cytokeratin 18 antibodies. Immunoblotting was performed as described in Supporting Informa-tion. The paper explained PROBLEM: While metastatic disease underlies most cancer-related mortality, few genetic lesions that select for metastatic tumour cell variants have been identified. Amoeboid motility is one of several diverse modes adopted by disseminating tumour cells. Elucidation of networks and critical signaling nodes that confer or restrain the amoeboid phenotype would facilitate discovery of novel therapies to control metastasis. The DIAPH3 locus, encoding the protein Diaphanous-related formin-3 (DIAPH3), resides at a chromosomal location that is frequently lost in metastatic prostate cancer. The potential functional significance of loss of this locus is unknown. RESULTS: Analysis of genome-wide, SCNA revealed that the DIAPH3 locus was a consensus area of chromosomal deletion common to several carcinomas. DIAPH3 deletions accumulated during disease progression, were strongly associated with metastatic disease, and were prevalent in DTCs from patient bone marrow aspirates. DIAPH3 silencing cooperated with oncogenic trans-formation to evoke an amoeboid phenotype in several tumour cell backgrounds. Loss of DIAPH3 caused cytoskeletal and endocytic trafficking defects through which EGFR/MEK/ERK signaling was hyperactivated. Pharmacologic inhibition of MEK suppressed the amoeboid phenotype, but tyrosine kinase inhibitors were ineffective. DIAPH3 silencing potentiated for-mation of pulmonary metastases in vivo, and its loss correlated with metastasis in human tumours. IMPACT: This is the first report showing that loss of a cytoskeleton remodelling protein, encoded by a locus that is lost at high frequency in multiple tumours and is strongly associated with metastasis, results in acquisition of the amoeboid cancer cell phenotype. These results may have prognostic utility to distinguish low-risk from high-risk disease. www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 757
  • 17. RNAi For sequences and transfection methods, see Supporting Information. Multiple, independent hairpins and duplexes (4) produced similar results in numerous readouts. Immunofluorescence Antibodies used: Acetylated tubulin and Rab11 (Abcam), FITC-conjugated anti-FLAG (Sigma), FITC-conjugated anti-EEA1 (BD Bios-ciences), EGFR (Biosource), FITC-Cholera Toxin B (CTxB, Sigma), FAK(Y397), MLC2, pMLC2(S19) and Rab5, (Cell Signaling). F-actin was detected with rhodamine-phalloidin (Invitrogen). An Axioplan 2 microscope (Zeiss) equipped with an AxioCam camera was used for fluorescence imaging. Quantitation of EGFR-positive endosomes and focal adhesions: Duplicate slides were evaluated by three observers blinded to the experimental group in two independent experiments, in 50 cells/ condition per experiment. Tubulin acetylation: DU145 cells were incubated with acetylated tubulin antibodies, and fluorescence quantified using Axiovision 4.5 software (Zeiss). Images were rendered using ImageJ processing software. Where noted, cells were stained with rhodamine–phalloidin or FITC-CTxB prior to fixation. Rab11 co-localization: DU145 cells were incubated with antibodies against Rab11 and EGFR, and fluorescent co-localization analysed with ImageJ. Regions with pixel intensity common to Rab11 and EGFR were highlighted. Where indicated, cells plated on collagen I were pretreated with nocodazole (2mM, Sigma) for 30 min, and processed as above with EGFR antibodies. Morphogenesis assay Three-dimensional culture of epithelial cells was performed as described (Debnath et al, 2002). Briefly, cells were seeded onto Matrigel basement membrane matrix, and acini size measured and analysed with Fiji image processing software. Luminescence proliferation assay Viable cells were quantified with the CellTiter-Glo assay (Promega), with luminescence measured every 24 h with a FLUOstar Omega platereader (BMG Labtech). Experimental Metastasis Model Animal studies were conducted in compliance with Children’s Hospital Boston IACUC guidelines using BALB/c nude mice (Massachusetts General Hospital). DU145 cells expressing control or DIAPH3 shRNAs were resuspended in HBSS, injected into the tail veins of anesthetized mice (15 animals per group), and after 8 weeks mice were sacrificed, lungs isolated and each lobe fixed individually in 10% formalin. Time-lapse video microscopy and track-plot analysis Cell migration was monitored with an inverted microscope, collecting images every 2 min for 30 h, and movies analysed using Bioimaging software (Andor). Real-time invasion assay Cells labelled with CellTracker Red CMTPX (Molecular Probes) were seeded onto collagen I-coated FluoroBlokTM inserts, and fluorescence intensity of cells migrated towards chemo-attractants monitored at 6 or 12 h. Statistical analyses Student’s t-test (two-tailed) was used if data were normally distributed. Otherwise, Mann–Whitney U, ANOVA, or Fisher’s exact tests were used. Analyses were performed with Prism 5.0a software. Data were plotted as meanSD. Author contributions MRF, MHH, and SM conceived the study; MHH, SM, DDV and MRF designed the experiments; MHH, SM, DDV, MM, KRS and JK performed experiments and analysed the data, including statistical analyses, with assistance from DRB, SG and GM; WL, FD, INH, WBI, HNH and RLV provided reagents, primary data sets, and bioinformatics analyses; MHH, SM, and MRF wrote the manuscript, with assistance by DDV and RMA. Acknowledgements The authors thank Joan S. Brugge and Wei Yang for helpful discussions, and Delia Lopez, Jiyoung Choi and Maosong Qi for technical assistance. Joseph Khoury and Mohini Lutchman made contributions in the study’s early phases. Grant support: NCI R01 CA143777, CA112303, NIDDK R3747556, P50 DK65298 and DAMD17-03-2-0033 (M.R.F.); US Department of Defense Prostate Cancer Research Program W81XWH-07-1- 0148 and AUAF/GlaxoSmithKline (M.H.H.); NIH R00 CA131472 (D.D.V.); American Institute for Cancer Research 09A107 (S.M.); and NCI R01 CA135008, CA133066 (W.L.). Supporting Information is available at EMBO Molecular Medicine online. The authors declare that they have no conflict of interest. References Bartolini F, Gundersen GG (2010) Formins and microtubules. Biochim Biophys Acta 1803: 164-173 Bartolini F, Moseley JB, Schmoranzer J, Cassimeris L, Goode BL, Gundersen GG (2008) The formin mDia2 stabilizes microtubules independently of its actin nucleation activity. J Cell Biol 181: 523-536 Belletti B, Nicoloso MS, Schiappacassi M, Berton S, Lovat F, Wolf K, Canzonieri V, D’Andrea S, Zucchetto A, Friedl P, et al (2008) Stathmin activity influences sarcoma cell shape, motility, and metastatic potential. Mol Biol Cell 19: 2003-2013 Beroukhim R, Getz G, Nghiemphu L, Barretina J, Hsueh T, Linhart D, Vivanco I, Lee JC, Huang JH, Alexander S, et al (2007) Assessing the significance of chromosomal aberrations in cancer: Methodology and application to glioma. Proc Natl Acad Sci USA 104: 20007-20012 Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, Barretina J, Boehm JS, Dobson J, Urashima M, et al (2010) The landscape of somatic copy-number alteration across human cancers. Nature 463: 899- 905 Berton S, Belletti B, Wolf K, Canzonieri V, Lovat F, Vecchione A, Colombatti A, Friedl P, Baldassarre G (2009) The tumor suppressor functions of p27(kip1) include control of the mesenchymal/amoeboid transition. Mol Cell Biol 29: 5031-5045 Research Article DIAPH3 and the amoeboid phenotype 758 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org
  • 18. Bignell GR, Greenman CD, Davies H, Butler AP, Edkins S, Andrews JM, Buck G, Chen L, Beare D, Latimer C, et al (2010) Signatures of mutation and selection in the cancer genome. Nature 463: 893-898 Burger M, Denzinger S, Hammerschmied C, Tannapfel A, Maderstorfer A, Wieland WF, Hartmann A, Stoehr R (2006) Mitogen-activated protein kinase signaling is activated in prostate tumors but not mediated by B-RAF mutations. Eur Urol 50: 1102-1109; discussion 1109–1110 Chalkia D, Nikolaidis N, Makalowski W, Klein J, Nei M (2008) Origins and evolution of the formin multigene family that is involved in the formation of actin filaments. Mol Biol Evol 25: 2717-2733 Debnath J, Mills KR, Collins NL, Reginato MJ, Muthuswamy SK, Brugge JS (2002) The role of apoptosis in creating and maintaining luminal space within normal and oncogene-expressing mammary acini. Cell 111: 29-40 Deribe YL, Wild P, Chandrashaker A, Curak J, Schmidt MH, Kalaidzidis Y, Milutinovic N, Kratchmarova I, Buerkle L, Fetchko MJ, et al (2009) Regulation of epidermal growth factor receptor trafficking by lysine deacetylase HDAC6. Sci Signal 2: ra84 Di Vizio D, Kim J, Hager MH, Morello M, Yang W, Lafargue CJ, True LD, Rubin MA, Adam RM, Beroukhim R, et al (2009) Oncosome formation in prostate cancer: Association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res 69: 5601-5609 Dong JT, Chen C, Stultz BG, Isaacs JT, Frierson HF, Jr (2000) Deletion at 13q21 is associated with aggressive prostate cancers. Cancer Res 60: 3880- 3883 Fernandez-Borja M, Janssen L, Verwoerd D, Hordijk P, Neefjes J (2005) RhoB regulates endosome transport by promoting actin assembly on endosomal membranes through Dia1. J Cell Sci 118: 2661-2670 Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, Chen H, Omeroglu G, Meterissian S, Omeroglu A, et al (2008) Stromal gene expression predicts clinical outcome in breast cancer. Nat Med 14: 518- 527 Friedl P, Wolf K (2010) Plasticity of cell migration: A multiscale tuning model. J Cell Biol 188: 11-19 Gaillard J, Ramabhadran V, Neumann E, Gurel P, Blanchoin L, Vantard M, Higgs HN (2011) Differential interactions of the formins INF2, mDia1, and mDia2 with microtubules. Mol Biol Cell 22: 4575-4587 Gao YS, Hubbert CC, Yao TP (2010) The microtubule-associated histone deacetylase 6 (HDAC6) regulates epidermal growth factor receptor (EGFR) endocytic trafficking and degradation. J Biol Chem 285: 11219-11226 Gasman S, Kalaidzidis Y, Zerial M (2003) RhoD regulates endosome dynamics through diaphanous-related formin and Src tyrosine kinase. Nat Cell Biol 5: 195-204 Gioeli D, Mandell JW, Petroni GR, Frierson HF, Jr, WeberMJ (1999) Activation of mitogen-activated protein kinase associated with prostate cancer progression. Cancer Res 59: 279-284 Goode BL, Eck MJ (2007) Mechanism and function of formins in the control of actin assembly. Annu Rev Biochem 76: 593-627 Hamadi A, Bouali M, Dontenwill M, Stoeckel H, Takeda K, Ronde P (2005) Regulation of focal adhesion dynamics and disassembly by phosphorylation of FAK at tyrosine 397. J Cell Sci 118: 4415-4425 Holcomb IN, Grove DI, Kinnunen M, Friedman CL, Gallaher IS, Morgan TM, Sather CL, Delrow JJ, Nelson PS, Lange PH, et al (2008) Genomic alterations indicate tumor origin and varied metastatic potential of disseminated cells from prostate cancer patients. Cancer Res 68: 5599-5608 Hosoki S, Ota S, Ichikawa Y, Suzuki H, Ueda T, Naya Y, Akakura K, Igarashi T, Oshimura M, Nihei N, et al (2002) Suppression of metastasis of rat prostate cancer by introduction of human chromosome 13. Asian J Androl 4: 131- 136 Huang DH, Su L, Peng XH, Zhang H, Khuri FR, Shin DM, Chen ZG (2009) Quantum dot-based quantification revealed differences in subcellular localization of EGFR and E-cadherin between EGFR-TKI sensitive and insensitive cancer cells. Nanotechnology 20: 225102 Joffre C, Barrow R, Menard L, Calleja V, Hart IR, Kermorgant S (2011) A direct role for Met endocytosis in tumorigenesis. Nat Cell Biol 13: 827-837 Research Article Martin H. Hager et al. Kainu T, Juo SH, Desper R, Schaffer AA, Gillanders E, Rozenblum E, Freas-Lutz D, Weaver D, Stephan D, Bailey-Wilson J, et al (2000) Somatic deletions in hereditary breast cancers implicate 13q21 as a putative novel breast cancer susceptibility locus. Proc Natl Acad Sci USA 97: 9603-9608 Kalluri R, Weinberg RA (2009) The basics of epithelial–mesenchymal transition. J Clin Invest 119: 1420-1428 Lanahan AA, Hermans K, Claes F, Kerley-Hamilton JS, Zhuang ZW, Giordano FJ, Carmeliet P, Simons M (2010) VEGF receptor 2 endocytic trafficking regulates arterial morphogenesis. Dev Cell 18: 713-724 Li HR, Wang-Rodriguez J, Nair TM, Yeakley JM, Kwon YS, Bibikova M, Zheng C, Zhou L, Zhang K, Downs T et al (2006) Two-dimensional transcriptome profiling: Identification of messenger RNA isoform signatures in prostate cancer from archived paraffin-embedded cancer specimens. Cancer Res 66: 4079-4088 Liu W, Laitinen S, Khan S, Vihinen M, Kowalski J, Yu G, Chen L, Ewing CM, Eisenberger MA, Carducci MA, et al (2009) Copy number analysis indicates monoclonal origin of lethal metastatic prostate cancer. Nat Med 15: 559- 565 Lizarraga F, Poincloux R, Romao M, Montagnac G, Le Dez G, Bonne I, Rigaill G, Raposo G, Chavrier P (2009) Diaphanous-related formins are required for invadopodia formation and invasion of breast tumor cells. Cancer Res 69: 2792-2800 Mas VR, Maluf DG, Archer KJ, Yanek K, Kong X, Kulik L, Freise CE, Olthoff KM, Ghobrial RM, McIver P, et al (2009) Genes involved in viral carcinogenesis and tumor initiation in hepatitis C virus-induced hepatocellular carcinoma. Mol Med 15: 85-94 Mosesson Y, Mills GB, Yarden Y (2008) Derailed endocytosis: An emerging feature of cancer. Nat Rev Cancer 8: 835-850 Nishimura Y, Bereczky B, Ono M (2007) The EGFR inhibitor gefitinib suppresses ligand-stimulated endocytosis of EGFR via the early/late endocytic pathway in non-small cell lung cancer cell lines. Histochem Cell Biol 127: 541-553 Peng J, Wallar BJ, Flanders A, Swiatek PJ, Alberts AS (2003) Disruption of the Diaphanous-related formin Drf1 gene encoding mDia1 reveals a role for Drf3 as an effector for Cdc42. Curr Biol 13: 534-545 Pinner S, Sahai E (2008) PDK1 regulates cancer cell motility by antagonising inhibition of ROCK1 by RhoE. Nat Cell Biol 10: 127-137 Sahai E, Marshall CJ (2003) Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol 5: 711-719 Sanz-Moreno V, Gadea G, Ahn J, Paterson H, Marra P, Pinner S, Sahai E, Marshall CJ (2008) Rac activation and inactivation control plasticity of tumor cell movement. Cell 135: 510-523 Sanz-Moreno V, Marshall CJ (2010) The plasticity of cytoskeletal dynamics underlying neoplastic cell migration. Curr Opin Cell Biol 22: 690-696 Schmidt S, Friedl P (2010) Interstitial cell migration: Integrin-dependent and alternative adhesion mechanisms. Cell Tissue Res 339: 83-92 Soldati T, Schliwa M (2006) Powering membrane traffic in endocytosis and recycling. Nat Rev Mol Cell Biol 7: 897-908 Sonee M, Barron E, Yarber FA, Hamm-Alvarez SF (1998) Taxol inhibits endosomal–lysosomal membrane trafficking at two distinct steps in CV-1 cells. Am J Physiol 275: C1630-C1639 Sorkin A, von Zastrow M (2009) Endocytosis and signalling: Intertwining molecular networks. Nat Rev Mol Cell Biol 10: 609-622 Stastna J, Pan X, Wang H, Kollmannsperger A, Kutscheidt S, Lohmann V, Grosse R, Fackler OT (2011) Differing and isoform-specific roles for the formin DIAPH3 in plasma membrane blebbing and filopodia formation. Cell Res 22: 728-745 Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, et al (2010) Integrative genomic profiling of human prostate cancer. Cancer Cell 18: 11-22 Thomas RK, Baker AC, Debiasi RM, Winckler W, Laframboise T, Lin WM, Wang M, Feng W, Zander T, MacConaill L, et al (2007) High-throughput oncogene mutation profiling in human cancer. Nat Genet 39: 347-351 Verhey KJ, Gaertig J (2007) The tubulin code. Cell Cycle 6: 2152-2160 www.embomolmed.org EMBO Mol Med 4, 743–760 2012 EMBO Molecular Medicine 759
  • 19. Wallar BJ, Deward AD, Resau JH, Alberts AS (2007) RhoB and the mammalian Diaphanous-related formin mDia2 in endosome trafficking. Exp Cell Res 313: 560-571 Wang Y, Roche O, Yan MS, Finak G, Evans AJ, Metcalf JL, Hast BE, Hanna SC, Wondergem B, Furge KA, et al (2009) Regulation of endocytosis via the oxygen-sensing pathway. Nat Med 15: 319-324 Wikman H, Vessella R, Pantel K (2008) Cancer micrometastasis and tumour dormancy. Apmis 116: 754-770 Wolf K, Mazo I, Leung H, Engelke K, von Andrian UH, Deryugina EI, Strongin AY, Brocker EB, Friedl P (2003) Compensation mechanism in tumor cell migration: Mesenchymal–amoeboid transition after blocking of pericellular proteolysis. J Cell Biol 160: 267-277 Wyckoff JB, Pinner SE, Gschmeissner S, Condeelis JS, Sahai E (2006) ROCK- and myosin-dependent matrix deformation enables protease-independent tumor–cell invasion in vivo. Curr Biol 16: 1515-1523 Research Article DIAPH3 and the amoeboid phenotype 760 2012 EMBO Molecular Medicine EMBO Mol Med 4, 743–760 www.embomolmed.org