ArticlePDF Available

Deciphering Tumour Tissue Organization by 3D Electron Microscopy and machine learning

Authors:

Abstract and Figures

Despite recent progress in the characterization of tumour components, the tri-dimensional (3D) organization of this pathological tissue and the parameters determining its internal architecture remain elusive. Here, we analysed the spatial organization of patient-derived xenograft tissues generated from hepatoblastoma, the most frequent childhood liver tumour, by serial block-face scanning electron microscopy using an integrated workflow combining 3D imaging, manual and machine learning-based semi-automatic segmentations, mathematics and infographics. By digitally reconstituting an entire hepatoblastoma sample with a blood capillary, a bile canaliculus-like structure, hundreds of tumour cells and their main organelles (e.g. cytoplasm, nucleus, mitochondria), we report unique 3D ultrastructural data about the organization of tumour tissue. We found that the size of hepatoblastoma cells correlates with the size of their nucleus, cytoplasm and mitochondrial mass. We also found anatomical connections between the blood capillary and the planar alignment and size of tumour cells in their 3D milieu. Finally, a set of tumour cells polarized in the direction of a hot spot corresponding to a bile canaliculus-like structure. In conclusion, this pilot study allowed the identification of bioarchitectural parameters that shape the internal and spatial organization of tumours, thus paving the way for future investigations in the emerging onconanotomy field.
This content is subject to copyright. Terms and conditions apply.
ARTICLE
Deciphering tumour tissue organization by 3D
electron microscopy and machine learning
Baudouin Denis de Senneville1,2,11, Fatma Zohra Khoubai 3,11, Marc Bevilacqua4, Alexandre Labedade 5,
Kathleen Flosseau6, Christophe Chardot7, Sophie Branchereau8, Jean Ripoche9, Stefano Cairo 6,10,12,
Etienne Gontier 4,12 & Christophe F. Grosset 3,12
Despite recent progress in the characterization of tumour components, the tri-dimensional
(3D) organization of this pathological tissue and the parameters determining its internal
architecture remain elusive. Here, we analysed the spatial organization of patient-derived
xenograft tissues generated from hepatoblastoma, the most frequent childhood liver tumour,
by serial block-face scanning electron microscopy using an integrated workow combining
3D imaging, manual and machine learning-based semi-automatic segmentations, mathe-
matics and infographics. By digitally reconstituting an entire hepatoblastoma sample with a
blood capillary, a bile canaliculus-like structure, hundreds of tumour cells and their main
organelles (e.g. cytoplasm, nucleus, mitochondria), we report unique 3D ultrastructural data
about the organization of tumour tissue. We found that the size of hepatoblastoma cells
correlates with the size of their nucleus, cytoplasm and mitochondrial mass. We also found
anatomical connections between the blood capillary and the planar alignment and size of
tumour cells in their 3D milieu. Finally, a set of tumour cells polarized in the direction of a hot
spot corresponding to a bile canaliculus-like structure. In conclusion, this pilot study allowed
the identication of bioarchitectural parameters that shape the internal and spatial organi-
zation of tumours, thus paving the way for future investigations in the emerging
onconanotomy eld.
https://doi.org/10.1038/s42003-021-02919-z OPEN
1CNRS, University of Bordeaux, Institut de Mathématiques de Bordeaux(IMB), UMR5251, 351 cours Libération, F-33400 Talence, France. 2INRIA Bordeaux,
MONC team, 200 av Vieille Tour, F-33400 Talence, France. 3Univ. Bordeaux, INSERM, Biotherapy of Genetic Diseases, Inammatory Disorders and Cancer,
BMGIC, U1035, MIRCADE team, 146 rue Léo Saignat, 33076 Bordeaux, France. 4Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Centre, BIC, UMS 3420,
US 4, F-33000 Bordeaux, France. 5Alexandre Labedade, freelance, F-33870 Vayres, France. 6XenTech Company, Genopole Campus 3, 4 Rue Pierre Fontaine,
F-91000 Évry-Courcouronnes, France. 7Chirurgie Pédiatrique-Transplantation, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 149
rue de Sèvres, F-75015 Paris, France. 8Paediatric Surgery and Liver Transplant unit, Hôpital Bicêtre, Assistance Publique Hôpitaux de Paris Université Paris
Saclay, 78 rue du Général Leclerc, F-94270 Le Kremlin-Bicêtre, France. 9Univ. Bordeaux, INSERM, retired, 146 rue Léo Saignat, 33076 Bordeaux, France.
10 Istituto di Ricerca Pediatrica, Corso Stati Uniti 4, 35127 Padova, Italy.
11
These authors contributed equally: Baudouin Denis de Senneville, Fatma Zohra
Khoubai.
12
These authors jointly supervised this work: Stefano Cairo, Etienne Gontier, Christophe F. Grosset. email: etienne.gontier@u-bordeaux.fr;
christophe.grosset@inserm.fr
COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio 1
1234567890():,;
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Diagnostic, prognostic and predictive clinical cancer studies
are mainly based on the analysis of tissue sections in two
dimensions (2D). However, tumours often present a
complex architecture that 2D imaging cannot capture. Tumour cells
establish composite interactions with the surrounding cancerous cells,
extracellular matrix and stromal components, including blood
capillaries and immune cells. In recent investigations, we noted that
various hepatoma cells engrafted on the chick embryo chorioallantoic
membrane (CAM) were arranged histologically and microscopically
in a lineage-specicwaywhilegrowinginacomparablecontrolled
environment1,2. Moreover, pathological tumour atlases display a
multitude of 2D images with tumour tissues whose cells are orga-
nized differently while belonging to the same category of cancer
(http://www.pathologyatlas.ro;https://www.proteinatlas.org;https://
atlases.muni.cz). These observations suggest that the structural
organization of tumour tissue changes from one sample to another
andislikelycontrolledbyenvironmental and genetic factors such as
the tissue origin, state of differentiation, genetic programme, the
mutations they harbour, the activated molecular pathways, and
the surrounding stromal components such as blood capillaries.
Moreover, these interactions are dynamic and vary as the cancer
progresses and metastasizes. To shed light on this critical issue, we
investigated the ultrastructural pattern of cancer tissue.
The applications of 3D electron microscopy (EM) are still
under development. Since its advent in the 1930s, EM has allowed
the in-depth analysis of a wide range of biological samples.
Transmission electron microscopy (TEM) and scanning electron
microscopy (SEM) are routinely used to collect ultrastructural
data from biological specimens at nanoscale resolution and to
correlate structural images with biological functions. Although
effective, these techniques produce images only in two dimen-
sions. Therefore, investigating a large volume (1001000 pL) of
biological tissue in 3D at a high resolution was hardly possible
until the development of 3D EM technologies including serial
block-face (SBF)-SEM and Focused Ion Beam-SEM36.
Hepatoblastoma (HB) is the most common form of liver cancer
in young children. Recently, we reported the classication of these
paediatric tumours in three groups, the use of Velcade®as a new
therapeutic option for the treatment of aggressive HB and the
development of an HB model in chick embryo for biological
studies1,7,8. We also developed HB-patient-derived xenografts
(PDX), a tumour model that recapitulates the histological, genetic
and biological characteristics of parental HB9. Here, we used SBF-
SEM to investigate the 3D internal organization of HB samples
obtained from HB PDXs. Following image acquisition by SBF-
SEM, we gathered quantitative data about the ultrastructure of
tumour tissue using 3D imaging tools, mathematical algorithms
and deep-learning approaches. We accurately measured the size
of tumour cells and their main subcellular components (e.g.
nucleus, mitochondria and cytoplasm), their planar alignment,
their spatial orientation and the distance between the cells and a
blood capillary. Results validate the relevance of our integrated
workow using wide-eld 3D EM and computational approaches
to investigate the deep internal architecture of HB tissue and the
spatial organization of the tumour cells. These structural and
functional data pave the way for future investigations in the
emerging eld of onconanotomy.
Results
Image acquisition by EM and SBF-SEM of HB PDX tissues.To
study the 3D organization of HB-PDX tissues, we adapted the
protocol described by Deerinck et al.10 to tumour samples
(Supplementary Fig. 1) and analysed the ultrastructural mor-
phology and the region of interest (ROI) of each stained sample
by TEM using ultrathin sections. Figure 1a shows that the
staining of the four samples was uniform and well contrasted and
that all HB tissues retained their structure of origin. For example,
membranes, mitochondria, endoplasmic reticulum, nuclei, lipid
droplets and circulating red blood cells into capillaries (bc) were
clearly identiable and typical with an excellent grey tone scale.
The morphology of endothelial and immune cells was typical.
Intercellular spaces were also visible and identied as bile
canaliculus-like structures (bi, sample 2)11,12, which are indi-
cative of liver-derived tissue. Overall, these results demonstrated
the preservation of the ultrastructure of our HB samples following
xation and staining procedures.
Next, we analysed two HB PDXs by SBF-SEM and acquired
high-resolution images in Xand Yalong the Z-axis to produce a
substantial volumetric view of each sample. To obtain both image
quality and detailed volumetric information, the acquired volume
Fig. 1 Analysis of HB PDXs by EM. a Analysis of four HB PDX by TEM at
different magnications. White dotted-line frame: Region-of-interest
corresponding to immediately higher magnication. bc, blood capillary; bi,
bile canaliculus-like structure; EC, endothelial cells; MA, macrophage; lv,
lipid vesicles; mb; plasma membrane; mi, mitochondrion; n, nucleus; ni,
nuclear invagination; nl, nucleolus; LY, lymphocyte; RC, red cells; TC,
tumour cell. bTop panels: SBF-SEM stacks from two samples. Bottom
panels: Extracted images of orthogonal plane from different faces of
reconstructed volume (XY,YZ,XZ), whose positions are displayed on
orthoslice view positions inside stack. Scale bar =10 µm.
ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z
2COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio
Content courtesy of Springer Nature, terms of use apply. Rights reserved
was about 70 µm × 70 µm × 25 µm (pixel time =10 µs, pixel size =
15 nm). After acquisition, the images were aligned to obtain 3D
stacks (Fig. 1b; Sample 1: 250 images, 113.9 pL; Sample 2: 246
images, 121.6 pL). While volumetric image resolution was not
isotropic (15 nm lateral resolution versus 100 nm depth resolution),
ultrastructural features within the 3D stack were perfectly
maintained and identiable, allowing us to visualize the cells and
their subcellular components within the tumour samples from any
angleofviewonthethreeaxes(Fig.1b).
3D organization of individual tissue components. Following
stack reconstruction, we analysed the tissue features in tumour
Sample 1 (Supplementary Fig. 2a, b, Supplementary Video 1).
Using Ilastik13 and VAST-Lite14 software, we manually seg-
mented a blood capillary and its circulating red and white blood
cells (one leucocyte and one structure evoking a dendritic cell or a
platelet cluster) (Supplementary Fig. 2c, f, Supplementary
Video 1). The volume of the blood capillary portion irrigating the
tissue was 11.3 pL and the 11 entirely segmented murine red
blood cells were 61.92 ± 16.26 µm3in size (Supplementary
Fig. 2cf, Supplementary Data 1 and Supplementary Video 1).
Next, we manually segmented a tumour cell from Sample 2 by
delimitating the cytoplasm, the nucleus and the mitochondria.
Again, the 3D digital representation allowed volumetric quanti-
cation of the cell and its cytoplasm, nucleus and mitochondrial
network (970.6, 657.1, 313.6 and 64.6 µm3, respectively), as well
as affording a morphologic view of the tumour cell in its 3D
environment (Supplementary Fig. 3; Supplementary Video 2).
The size of this tumour cell was consistent with the size range of
HB cells15,16.
Reconstruction of tumour 3D organization by semi-automated
segmentation. Although it produces accurate data, manual seg-
mentation is too laborious and time-consuming to allow the
analysis of hundreds of cells constituting a stack of about 110120
pL. To overcome this problem and anticipate the analysis of
larger volumes of tumour (>200 pL), we implemented a semi-
automatic segmentation procedure comprising the manual seg-
mentation of the cytoplasm and nucleus of cells on 1 out of 10
images in the stack form Sample 2 and an automatic segmenta-
tion algorithm of ROI by proximal propagation along the Zaxis
(Supplementary Fig. 4). This procedure allowed us to segment
partially and entirely 182 tumour cells, 113 nuclei, 1 immune cell
and 3 portions of the same blood capillary (Fig. 2ac, Supple-
mentary Fig. 5, Supplementary Video 3). The elongated shape of
the immune cell, the presence of many vacuoles in its cytoplasm
and its location near the blood capillary may suggest of a tumour-
inltrating macrophage (Supplementary Fig. 5, Supplementary
Video 3). Following the alignment in a plane of 47 tumour cells
with a complete nucleus, we found that 36 tumour cells (76.6%)
and 35 nuclei (74.5%) had an inclination angle of 0 to 20°
(relative to the best alignment plane, see Online Methods), while
the inclination angle of the blood capillary was 8° (Fig. 2d, e,
Supplementary Fig. 6, Supplementary Data 1, Supplementary
Video 4). These data suggested that the blood capillary may
inuence the spatial arrangement of tumour cells.
Next, we measured the tumour cell polarity (in the sense
of attraction toward a particular object) using a vectoral approach
(see Online Methods). In both cases, data showed that a set
of tumour cells polarized in the direction of a hot spot
corresponding to a bile canaliculus-like structure (Fig. 3,
Supplementary Fig. 7, Supplementary Video 5)11,12. Numerous
membrane protrusions were visible at the boundary between
the bile canaliculus-like structure and the canalicular membrane
of the surrounding tumour cells (Supplementary Fig. 7,
Supplementary Video 5). This structure into which hepatocytes
excrete their metabolic agents after enzymatic neutralization is
specic to normal liver tissue15 and has been reported to be
remnant in some HB11,12.
Finally, we focused our attention on the 21 fully segmented
tumour cells contained in sample 2. Mitochondria are key
organelles of tumour cell function but too numerous (between
200 and 400 per cell) and small in size to be manually segmented
from our whole tissue sample. Thus, a deep-learning algorithm
was fed using data from mitochondria manually segmented on
one single cell. Next, we performed two successive cycles of
automatic segmentation by deep-learning propagation combined
with manual segmentation clean-up of segmented mitochondria
(see Methods). Following this segmentation procedure, the size of
these cells and their cytoplasm, nucleus and mitochondrial
network was measured and component/cell ratios were calculated
(Supplementary Fig. 8a, b, Supplementary Data 1). In line with
the known relationship between the size or number of organelles
and the size of the cell17,18, we found that the larger the tumour
cell, the larger its cytoplasm, global mitochondrial network and
nucleus (Fig. 4a, Supplementary Data 1). By measuring the
distance between each of these cellular components and the blood
capillary, we also found that the tumour cells located near the
blood capillary were signicantly larger than those located away
from it (Fig. 4b, c, Supplementary Data 1, Supplementary
Video 6). This inverse correlation with the distance to the
capillary was also observed when considering the cytoplasm and
the mitochondrial network (the latter having the highest
correlative value) but not the nuclei (Fig. 4c, Supplementary
Fig. 9ac, Supplementary Data 1, Supplementary Video 6).
Altogether, these data suggested that the size of HB cells and their
mitochondrial content is linked to the distance with blood
capillary. These observations are in agreement with the decrease
in mitochondrial mass in cells exposed to low oxygen supply19,20.
Discussion
A still unanswered question is whether cells are distributed ran-
domly in cancer tissue or if and how their organization is gov-
erned by physical and molecular factors reminiscent of the
structure of normal tissue. In this pilot study, we used a 3D EM
approach named onconanotomy, i.e. a workow to analyse in
depth the architecture of tumour samples using a combination of
sample preparation, SBF-SEM imaging, computational approa-
ches and infographic tools (see Technical workow in Fig. 5). To
the best of our knowledge, this is the rst report investigating the
complete ultrastructure of human tumour xenograft tissue by
high-resolution 3D EM.
While automated segmentation with machine learning meth-
ods has become an essential technique for analysing the ultra-
structure of all tissue and cell types, the main difculty in our
work laid in the construction of the learning database. At the very
beginning of our study, we did not have any organelle segmen-
tation available and we used manual segmentation. Thus, we
developed semi-automatic approaches to accelerate the process.
One can anticipate that a fully automated segmentation of major
organelles (cytoplasm, nuclei, mitochondria, etc.), cell types
(blood cells, immune cells, hepatocytes and tumour cells) and
tissue structures (capillary, bile-canaliculus, etc.) is feasible (see
Segmented tissue elements in Fig. 5) based on sufciently
populated training databases and given high potential of novel
technologies in articial intelligence.
Previous detailed reports on the 3D bioarchitectural organi-
zation of tumour are lacking to fully establish if our data are
reecting the real situation found in this pathological tissue.
However, we found red and white cells circulating in a blood
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio 3
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Fig. 2 Alignment of HB cells and nuclei. a Sample 2 stack. b,cDigital representation of (a) with 182 cells (b) and 113 nuclei (c). dAlignment of cells and
blood capillary in a plane: (i) coloured sticks =main axis of tumour cells; red stick =main axis of blood capillary. (iiix) Angles between each main cell axis
and the best alignment plane. 2D cross-sectional maps for increasing depth along Z-axis. Scale bar =10 µm. bdBlood capillary portions are in red.
eHistograms of cells (left panel) and nuclei (right panel) alignment angles. Red dashed line: alignment angle of blood capillary.
ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z
4COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Fig. 3 Tumour cell cluster with polarized shape orientation. Only tumour cells with a complete nucleus are considered here. Blood capillary portions are in
red. aAccumulation map of virtual rays [Accumulated ray intensity [a.u]; voxel-by-voxel basis; see yellowish voxels] emitted by each cell along its main
axis. (i) Reconstructed 3D image of accumulation map of virtual rays. (iiix) 2D cross-sectional maps for increasing depth along the Z-axis. bBinary
classication of polarised/unpolarised cells: (i) Reconstructed 3D image of polarised/unpolarised cells. 17 cells (in yellow) emitted virtual rays reaching the
main accumulation region observable in panel (vii) (accumulated ray intensity >3.5). a.u., arbitrary unit. (iiix) 2D cross-sectional maps for increasing
depth along the Z-axis. Scale bar =10 µm.
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio 5
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Fig. 4 3D organization of typical cellular and subcellular structures. Only 21 entirely contained cells are considered here. aVolumetric correlations
between cytoplasm, nucleus and mitochondrial network. bCell sizes related to distance from blood capillary: (i) Reconstructed 3D image of cells and blood
capillary. Cell colour is related to its volumetric size. (iiix) 2D cross-sectional maps for increasing depth along the Z-axis. (iix) Blood capillary portions are
in red. Scale bar =10 µm. cCorrelation between distance to blood capillary and cellular/subcellular structures or nucleocytoplasmic ratio. a,cSpearman
correlations. rand pvalues are as indicated in corresponding graph.
ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z
6COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio
Content courtesy of Springer Nature, terms of use apply. Rights reserved
capillary portion (Supplementary Fig. 2, Supplementary Video 1),
a direct correlation between the size of tumour cells, of mito-
chondria and of nuclei (Fig. 4a) and tumour cells polarizing in the
direction of a hot spot corresponding to a bile canaliculus-like
structure (Fig. 3, Supplementary Video 5). Together, these data
suggest that our samples preparation fully preserved the HB
sample ultrastructure following xation and staining procedures,
and consequently, that our structural observations are reliable.
Our data also demonstrate the feasibility of our methodology to
study the 3D organization of tumoral tissue using specic
bioarchitectural parameters (Fig. 5) and the potential advance it
represents in understanding cancer biology. The information
obtained on the organization of liver cancer cells throws light on
tumour physiology. In particular, it suggests that blood capillaries
could determine the ultrastructure of tumoral tissue by control-
ling the alignment and size of tumour cells and their subcellular
components. However, further studies are required to strengthen
this hypothesis. In addition, we found that a bile canaliculus-like
structure guides the spatial arrangement and the polarity of
tumour cells in HB tissue, as it does in normal liver tissue to
subsume the physiological functions of normal hepatocytes15.
This study is a preliminary attempt to unravel the internal
architecture of tumoral tissue and has limitations. First, it is
difcult to automatically segment-specic structures such as
tumour-inltrating macrophages and endoplasmic reticulum.
The procedure requires visual control and manual correction,
which are time-consuming. Second, our sample set was small.
Future studies with a larger number and variety of tumoral tissues
would allow us to generate more digitalized samples that in turn
would improve the automatic segmentation process. A larger
dataset would also allow us to better evaluate whether blood
capillaries and bile canaliculus-like structures, which are common
features in HB and more generally in epithelial liver tumours,
inuence the polarity and spatial organization of tumour cells and
their organelles. These techniques coupled with current omic
approaches would provide much insight into the relationship
between the structural organization of tumours, and their cellular
functions and metabolic activities.
Future possible directions of onconanotomy-based exploratory
studies are the following: (a) analysing virtually all types of solid
tumours to generate a virtual 3D biorepository whose data would
be particularly valuable in the case of rare tumours or precious
samples such as biopsies; (b) comparing matched tumour samples
such as changes occurring before and after treatment or primary
versus metastatic features, and (c) identifying structural subtypes
and verifying their overlap with different histological groups such
as embryonal versus foetal versus small undifferentiated cell
subtypes described in HB8,16. Since this methodology is adapted
to lab tumour models like cell xenografts, tumour-like spheroids
and organoids, our combined approach could help investigators
to clarify the role of genes and molecular pathways involved in
the architecture of tumoral tissue and to investigate the response
of tumour cells to drugs (e.g. checkpoint inhibitors, monoclonal
antibodies) and treatments (e.g. radiotherapy). Finally, our
approach is perfectly transferable to other biological kingdoms
and elds based on tissue development including for instance
plants, fungi, neuroscience, embryogenesis and biomaterials.
Based on our approach (Fig. 5), 3D EM users could measure
valuable structural parameters (e.g. size, component ratio,
polarity, etc.) and potential additional ones (e.g. cellular density,
surface, texture, etc.) related to tumour tissue components using
biopsies, tumour resections or lab models4. These analyses should
help investigators (1) to better understand how a tumour tissue is
spatially constructed, (2) to determine in a dynamic or com-
parative manner (e.g. at different tumour development periods)
the close relationships between tumour cells and other tissue
Fig. 5 Graphical summary of the 3D EM approach used to study tumour tissue architecture. The gure is divided into three steps: the technical workow,
the segmented tissue elements and the different bioarchitectural parameters used to analyse the 3D organization of tumour tissue.
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved
elements (e.g. blood capillary, nerve endings, immune cells, etc.)
and (3) to study in 3D the elements constituting a tumour
niche21. Such investigations could also help to clarify the
mechanisms by which the tissue is expanding while growing and
in which direction, how tumour cells invade the surrounding
tissue by outcompeting with non-tumoral cells and how the
metastatic cells traverse the vascular wall. By performing com-
parative analysis, investigators could evaluate at a nanoscale level
the efcacy of an antitumoral drug transported by the blood
circulation on tumour cells. The implementation of our approach
to histopathology practice may also provide additional bio-
markers for the diagnosis, the prognosis, the drug-response pre-
diction and help to stratify tumours. To sum up, our procedure
should boost the interest of the scientic and clinical commu-
nities working in cancer for 3D EM technologies4.
In conclusion, our study aims to foster the development of
what we have termed onconanotomy, a novel research eld in
oncology, that will complement other innovative imaging tech-
nologies such as two-photon excitation microscopy (also known
as 2PEF) and correlative light-electron microscopy (also known
as CLEM). This eld could be backed up by the development of
open-source databanks gathering all 3D imaging data at a
worldwide level in order to boost research and promote coop-
eration in cancer, as was previously the case with Gene databanks
and Cell and Tissue atlases. By accessing 3D image databanks,
investigators could upload valuable information and perform
comparative investigations to unravel the ultrastructure of solid
tumours. The potential focuses include cell-cell and cell-
extracellular matrix interactions, intratumoral heterogeneity,
pro-metastatic mechanisms, angiogenesis, drug response, ther-
apeutic efcacy and regulatory factor gradation. We believe that,
in few decades from now and thanks to digital approaches and
articial intelligence, these approaches could enrich the eld of
oncology with the support of other technologies such as single-
cell RNA-seq, microuidics and complex 3D lab models. It might
especially provide ground-breaking insights into how cells dis-
seminate and how drugs reach the targets cells. A better com-
prehension of the organization of tumour cells and clinical
outcome could help in identifying the key structural parameters
of cancers. In turn, this could lead to substantial advances in both
the understanding of cancers and the treatments of adult and
paediatric cancer patients.
Methods
Ethical clearance. PDX development program was approved by the Hospital ethics
committee, each sample was implanted upon signature of the related informed
consent form. Mice were maintained in specic pathogen-free animal housing at
the Center for Exploration and Experimental Functional Research (CERFE, Evry,
France) animal facility. All experimental procedures related to PDX development
were conducted in accordance with French regulatory legislation concerning the
protection of animals used for scientic purposes were approved by the CERFE
animal facility ethics committee and by the Ministère de lAgriculture et de lAli-
mentation, France.
PDX establishment. At surgery, tumour fragments were sampled from the
resected tumour and placed in cell culture medium supplemented with penicillin/
streptomycin and with or without 5% BSA on ice. Tumour samples were chopped
into 4 × 3 mm fragments and grafted in the interscapular region of 6-8 week-old
female athymic nude mice (Athymic Nude-Foxn1nu, ENVIGO-Harlan Labora-
tories, Gannat, France). Tumour growth from the rst implantation occurred with
a delay spanning between 1 and 5 months. Growing tumours were serially trans-
planted onto recipient mice and underwent comparative examination to conrm
preservation of their histological features. To immortalize each PDX, vials of 4 ×
3 mm fragments from tumours at different passages were placed in a solution of
90% FCS/10% DMSO or glycerol, and stored at 150 °C.
PDX samples. PDX was generated in compliance with the informed consent form
signed by the patients and developed as previously described9. The main clinical
and genetic characteristics of HB PDX samples are shown in Supplementary
Table 1.
PDX fragments xation. For each model, 5 fragments (2 × 1 mm) were taken and
placed in ice-cold xing solution (2% PFA and 2.5% glutaraldehyde in 0.15 M
Cacodylate +2 mM CaCl
2
buffer). After 3-h xation with gentle stirring, the
fragments were transferred to a tube containing Cacodylate buffer and kept
overnight at 4 °C before shipment to the Imaging platform.
Serial block-face scanning electron microscopy sample preparation. Tissue was
prepared for SBF-SEM as previously described10. The samples were xed with a 2%
paraformaldehyde and 2,5% solution of glutaraldehyde in 0.15 M cacodylate buffer
(pH 7.4) for 2 h at room temperature and then were washed 5 × 3 min in cold
0.15 M cacodylate buffer. En bloc contrast staining was performed by consecutive
incubations in heavy metal containing solutions. The rst staining step was a 1 h
incubation on ice in 2% OsO
4
containing 1.5% potassium ferrocyanide in 0.15 M
cacodylate buffer. After washing 5 × 3 min in ultrapure water, the samples were
incubated for 20 min in a fresh thiocarbohydrazide solution (1% w/v in water) at
room temperature. The next wash step was followed by incubation in 2% osmium
in water at RT for 30 min and after washing 5 × 3min in ultrapure water 2% uranyl
acetate at 4 °C overnight. The following day, Waltons lead aspartate staining was
performed for 30 min at 60 °C. For this, a 30 mM L-aspartic acid solution was used
to freshly dissolve lead nitrate (20 mM, pH 5.5), the solution was ltered and blocks
incubated for 30 min at 60 °C. After nal washing steps, the samples were dehy-
drated using ice-cold solutions of 30%, 50%, 70%, 90%, 2×100% ethanol (anhy-
drous), 2 × 100% aceton, 10 min each. Resin embedding was done using Epon by
rst placing the samples in 25% aceton/Epon for 2 h, 50% aceton/Epon for 2 h, 75%
aceton/epon for 2 h and followed by 2 incubations in 100% epon (overnight, 8 h).
The samples were put in fresh epons resin and placed at 60 °C for 48h. Once the
resin block were hardened, they were roughly cut with a razor blade to generate a
pyramid shape and mounted on aluminium specimen pins using a silver lled
conductive resin (Epotek-Delta microscopies, Mauressac, France). After 24 h of
polymerisation at 60 °C, the samples were trimmed with a diamond knife (Dia-
tome, Nidau, Switzerland). Silver lled conductive resin was used to electrically
connect the edges of the tissue to the aluminium pin. The entire sample was then
sputter coated with a 510 nm layer of gold to enhance conductivity.
Transmission electron microscopy. The samples were rst analysed by TEM to
control morphology and dene the region of interest. Ultra-thin sections were cut
and deposited on copper grids and observed with a Hitachi H7650 transmission
electron microscope (Japan).
Serial block-face scanning electron microscopy imaging. The sample on the pin
was placed into the carrier that ts into the3View ultramicrotome (3viewXP2-
Gatan Inc., Pleasanton, CA, USA) on a ZEISS Gemini eld emission gun SEM300
(Zeiss - Marly-le-Roi - France). The block face was imaged with an accelerating
voltage at 1.2 kV using the Back scattered electron with a specic BSE Detector (On
point - Gatan Inc., Pleasanton, CA, USA).
Pre-processing and structure segmentation. Image analysis was performed
using three complementary software packages. Digital Micrograph (Gatan Inc.,
Pleasanton, CA, USA) was used to align the images with the Image Alignment
plugin using the combined default lter (which combines soft rectangle and
bandpass lters). The aligned images were saved in Gatan format dm4as a single
stack. Fiji software was used to convert the 32-Bit images into 8-Bit images to be
less resource-consuming and facilitate segmentation. Then the image was cropped
to the optimal (larger) rectangular eld of view, and the brightness and contrast
were automatically adjusted (Z-scoring on a slice-by-slice basis). Finally, VAST-
Lite software was used for the manual segmentation of the elements of interest
(cells, nuclei and mitochondria)14. Image processing was done following the
pipeline shown in Supplementary Fig. 10. All manual segmentation was done using
an interactive drawing tablet and pen (Cintiq pro 5, Wacom).
The manual segmentation of both cells and nuclei was done only on 10% of the
stack of images (1 image out of 10) using the VAST-Lite software14. These manual
segmentations were propagated to neighbouring slices using a so-called « Optical
Flow » algorithm (see Source codessection below) applied on the EM images,
similar to the approach described by Huang T.C. et al.22. This procedure allowed us
to segment partially and entirely 182 tumour cells, 113 nuclei in a relatively decent
time frame (several weeks were mandatory). However, the segmentation of
mitochondria turned out to be a different challenge. The reason is two-fold: (1) the
volume of mitochondria is far lower the size of cells nuclei, preventing the use of
the above-mentioned propagation strategy; (2) the number of mitochondria is
much greater than the number of cells and nuclei, further hampering in turn the
amount of work. Mitochondria were thus segmented manually on one single cell to
feed a deep-learning algorithm (using a 3D U-net architecture23 and an
implementation based on Tensorow 1.4 and Keras 2.2.4), which could be
subsequently used for semi-automatic segmentation. The 3D U-Net architecture
used for the segmentation of mitochondria is presented in Supplementary Fig. 11.
The architecture is similar to the one proposed by Huo et al.24. We used one single
input channel (i.e., the SBF-image). The loss function was a combination of
categorical cross-entropy and Dice25. The optimizer algorithm was Adam with
default parameters26. 100 epochs were performed and the batch size was equal to 1.
ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z
8COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Due to limited GPU memory, we used a patch-wise strategy27: practically, local
patches of 128 × 128 × 128 voxels were extracted in the training SBF image and
used as inputs for the CNN. To nish, erroneous automatic segmentations of
mitochondria were corrected manually using the VAST-Lite14 and Ilastik13
software.
Analysis of orientation of structures. Each cell, nucleus and capillary portion was
characterized using its main axis. To this end, a Principal Component Analysis
(PCA) was applied on the voxel coordinates within the segmented mask of a
structure of interest. Only tumour cells with a complete nucleus were considered.
The following two analyses were conducted.
Analysis of alignment in 2D plane of cells and nuclei. The main axes of cells were
used to calculate the best 2D alignment plane(in the least-squares sense: we
minimized the sum of squared differences between the observed main axes of cells,
and the tted value provided by a 2D plane equation). Angles between the main
axis of each cell and the alignment plane were calculated. The same analysis was
conducted subsequently for nuclei.
Determination of cell clusters with polarized shape orientation. 3D virtual rays were
emitted by each cell along its main axis in both directions (the ray radius was a
user-dened input parameter for the algorithm and was set to 15 µm, ray intensity
proportional to the Euclidean distance to the main axis, with a value of 1 on the
main axis). To this end, Bresenhams line algorithm28 was employed and adapted
to our needs. The accumulated beam intensity was calculated on a voxel-by-voxel
basis. Cells emitting virtual rays reaching a specic accumulation region were
subsequently selected. Thus, cells converging toward a common region could be
listed.
Source codes. The code designed to identify the analyzed bioarchitectural para-
meters (alignment, volume, shape orientation) can be found in https://github.com/
bsenneville/Onconanotomy/. The code implementing the above-mentioned optical
ow algorithm employed for the semi-automatic segmentation of cells and nuclei
can be found in https://github.com/bsenneville/2D_Optical_Flow/29. Both codes
were developed under the commercial software Matlab ©1994-2021 The
MathWorks, Inc.
Infographics. Blender version 2.90.1 (https://www.blender.org) was used to create
the illustrations (images and Supplementary videos). The Cycles render engine was
used for all the nal renderings. To render polygonal meshes, the data were
imported from VTK les using the BVTK addon (https://github.com/tkeskita/
BVtkNodes). Depending on the data complexity, the meshes were decimated to
lower the polygon count, but still preserving the overall shape and most of the
details. For volumetric rendering, the original stack data was converted from a
multi-page Tiff image le to a downscaled 2D tile map, which was then loaded and
translated to a 3D volume into Blender using a custom shader setup. ImageJ
(https://imagej.nih.gov/ij/), XNview (https://www.xnview.com) and the montage
command line tool of ImageMagick (https://imagemagick.org/script/montage.php)
were used for image conversion, downscaling and stitching. Downscaling is
mandatory, since the original amount of data is not always practical to work with
and is not necessary for the purpose of visualization.
Statistical analyses. Statistical analyses were performed using GraphPad Prism
7.05 software. Spearmans nonparametric correlation was used to compare the size
of different cell components or the size of a specic component to the distance to a
blood capillary. Results were considered signicant when p< 0.05.
Reporting summary. Further information on research design is available in the Nature
Research Reporting Summary linked to this article.
Data availability
The data that support the ndings of this study are available within the paper and
its Supplementary information les.
Received: 17 August 2021; Accepted: 12 November 2021;
References
1. Indersie, E. et al. MicroRNA therapy inhibits hepatoblastoma growth in vivo
by targeting beta-catenin and Wnt signaling. Hepatol. Commun. 1, 168183
(2017).
2. Cartier, F. et al. New tumor suppressor microRNAs target glypican-3 in
human liver cancer. Oncotarget 8, 4121141226 (2017).
3. Denk, W. & Horstmann, H. Serial block-face scanning electron microscopy to
reconstruct three-dimensional tissue nanostructure. PLoS Biol. 2, e329 (2004).
4. Riesterer, J. L. et al. A workow for visualizing human cancer biopsies using
large-format electron microscopy. Methods Cell Biol. 158, 163181 (2020).
5. Knott, G., Marchman, H., Wall, D. & Lich, B. Serial section scanning electron
microscopy of adult brain tissue using focused ion beam milling. J. Neurosci.
28, 29592964 (2008).
6. Titze, B. & Genoud, C. Volume scanning electron microscopy for imaging
biological ultrastructure. Biol. Cell 108, 307323 (2016).
7. Indersie, E. et al. Tracking cellular and molecular changes in a species-specic
manner during experimental tumor progression in vivo. Oncotarget 9,
1614916162 (2018).
8. Hooks, K. B. et al. New insights into diagnosis and therapeutic options for
proliferative hepatoblastoma. Hepatology 68,89102 (2018).
9. Nicolle, D. et al. Patient-derived mouse xenografts from pediatric liver cancer
predict tumor recurrence and advise clinical management. Hepatology 64,
11211135 (2016).
10. Deerinck, T. J., Bushong, E., Thor, A. & Ellisman, M. H. NCMIR methods for
3D EM: a new protocol for preparation of biological specimens for serial
blockface scanning electron microscopy. Microscopy 1,68 (2010).
11. Dekmezian, R., Sneige, N., Popok, S. & Ordonez, N. G. Fine-needle aspiration
cytology of pediatric patients with primary hepatic tumors: a comparative
study of two hepatoblastomas and a liver-cell carcinoma. Diagn. Cytopathol. 4,
162168 (1988).
12. Pietsch, T. et al. Characterization of the continuous cell line HepT1 derived
from a human hepatoblastoma. Lab Invest 74, 809818 (1996).
13. Berg, S. et al. ilastik: interactive machine learning for (bio)image analysis. Nat.
Methods 16, 12261232 (2019).
14. Berger, D. R., Seung, H. S. & Lichtman, J. W. VAST (volume annotation and
segmentation tool): efcient manual and semi-automatic labeling of large 3d
image stacks. Front Neural Circuits 12, 88 (2018).
15. Cullen, J. M. & Stalker, M. J. in Jubb, Kennedy & Palmers Pathology of
Domestic Animals Vol. 2 (ed Grant Maxie, M.) 258352.e251 (2016).
16. Lopez-Terrada, D. et al. Towards an international pediatric liver tumor
consensus classication: proceedings of the Los Angeles COG liver tumors
symposium. Mod. Pathol. 27, 472491 (2014).
17. Chan, Y. H. & Marshall, W. F. Scaling properties of cell and organelle size.
Organogenesis 6,8896 (2010).
18. Schmoller, K. M. The phenomenology of cell size control. Curr. Opin. Cell
Biol. 49,5358 (2017).
19. Fuhrmann, D. C., Wittig, I., Heide, H., Dehne, N. & Brune, B. Chronic
hypoxia alters mitochondrial composition in human macrophages. Biochim.
Biophys. Acta 1834, 27502760 (2013).
20. Lages, Y. M. et al. Low oxygen alters mitochondrial function and response to
oxidative stress in human neural progenitor cells. PeerJ 3, e1486 (2015).
21. Laplane, L., Duluc, D., Larmonier, N., Pradeu, T. & Bikfalvi, A. The multiple
layers of the tumor environment. Trends Cancer 4, 802809 (2018).
22. Huang, T. C. et al. Semi-automated CT segmentation using optic ow and
Fourier interpolation techniques. Comput Methods Prog. Biomed. 84, 124134
(2006).
23. Ronneberger, O., Fischer, P. & Brox, T. U-net: Convolutional Networks for
Biomedical Image Segmentation. In International Conference on Medical
Image Computing and Computer-Assisted Intervention 234241 (Springer,
Cham, 2015) https://doi.org/10.1007/978-3-319-24574-4_28.
24. Huo, Y. et al. 3D whole brain segmentation using spatially localized atlas
network tiles. Neuroimage 194, 105119 (2019).
25. Sugino, T. et al. Loss weightings for improving imbalanced brain structure
segmentation using fully convolutional networks. Healthcare (Basel) 9,https://
doi.org/10.3390/healthcare9080938 (2021).
26. Kingma, D. P. & Ba, J. Adam: A Method for Stochastic Optimization. CoRR
abs/1412.6980 (2015).
27. Liu, Y., Ren, Q. R., Geng, J. H., Ding, M. & Li, J. Y. Efcient patch-wise
semantic segmentation for large-scale remote sensing images. Sensors-Basel
18, 3232 (2018).
28. Bresenham, J. Algorithm for computer control of a digital plotter. IBM Syst. J.
4,2530 (1965).
29. Zachiu, C., Papadakis, N., Ries, M., Moonen, C. & Denis de Senneville, B. An
improved optical ow tracking technique for real-time MR-guided beam
therapies in moving organs. Phys. Med Biol. 60, 90039029 (2015).
Acknowledgements
This work was supported by the charity Eva pour la Vie, La Fondation ARC pour la
Recherche sur le Cancer (contract N° PJA 20191209631), La Région Nouvelle-Aquitaine,
La Fondation Groupama pour la Santé and Groupama Centre-Atlantique. Microscopy
Imaging was performed at the Bordeaux Imaging Centre, which is a member of the
FranceBioImaging national infrastructure (ANR-10-INBS-04). Computational analysis
was carried out using the PlaFRIM experimental testbed, supported by Inria, CNRS
(LABRI and IMB), Université de Bordeaux, Bordeaux INP and Conseil Régional
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved
dAquitaine (see https://www.plafrim.fr/). Computer time for this study was provided by
the computing facilities at MCIA (Mésocentre de Calcul Intensif Aquitain) of the Uni-
versité de Bordeaux and of the Université de Pau et des Pays de lAdour.
Author contributions
Fresh paediatric tumour fragments were provided by S.B. and C.C. HB-PBX were
developed by XenTech company and provided by K.F. and S.C. Image acquisition and
analyses were performed by M.B., F.Z.K. and E.G. The manual segmentations and clean-
up of tissue and cellular components were performed by F.Z.K., M.B. and C.F.G. B.D.d.S.
performed the mathematical and computational analyses. A.L. generated 3D images and
videos. C.F.G., S.C. and E.G. supervised the work. C.F.G. obtained the nancial grants.
All authors actively participated in the writing of the manuscript.
Competing interests
The authors declare no competing interests.
Additional information
Supplementary information The online version contains supplementary material
available at https://doi.org/10.1038/s42003-021-02919-z.
Correspondence and requests for materials should be addressed to Etienne Gontier or
Christophe F. Grosset.
Peer review information Communications Biology thanks the anonymous reviewers for
their contribution to the peer review of this work. Primary Handling Editors: Chao Zhou
and Karli Montague-Cardoso.
Reprints and permission information is available at http://www.nature.com/reprints
Publishers note Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this license, visit http://creativecommons.org/
licenses/by/4.0/.
© The Author(s) 2021
ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02919-z
10 COMMUNICATIONS BIOLOGY | (2021) 4:1390 | https://doi.org /10.1038/s42003-021-02919-z | www.nature.com/commsbio
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Advances in 3D imaging technologies, such as serial block face-scanning electron microscopy (SBF-SEM) and focused ion beam-scanning electron microscopy (FIB-SEM), significantly improves the ability to characterize cellular components within biological tissues in 3D [1]. These techniques provide crucial insights into the fundamental principles underlying tissue spatial organization [2]. ...
... Intercellular segmentations were generated using a nnU-Net algorithm trained on manually annotated data [1], [5]. The obtained intercellular masks were then refined using a specialized neural network (COp-Net), as described in [7]. ...
... A 3D seeded watershed algorithm [9] was employed to roughly assign an individual label to each cell. The seeds were the cell nuclei segmented using a nnU-Net algorithm trained on manually annotated data [1]. ...
Preprint
Full-text available
Accurate 3D semantic cell segmentation in volumet-ric electron microscopy images is crucial for analyzing tissue architecture and plays a key role in studying fundamental biological processes. While current 2D semantic segmentation methods benefiting from numerous pre-trained networks available in the literature are effective, extending these approaches to 3D segmentation remains challenging. This study introduces a novel automated approach for enhancing 3D semantic cell segmentation by propagating 2D cell delineations and integrating intercellular priors to constrain segmentation within cell boundaries. We demonstrate the efficacy of our approach for automated 3D semantic cell segmentation using patient-derived xenograft (PDX) hepatoblastoma tissue samples acquired by serial block-face scanning electron microscopy. The proposed method significantly improves 3D semantic cell segmentation, paving the way for bioarchitectural investigations at the cellular level using biological and pathological samples.
... Scanning electron drug resistance. Therefore, the analysis of bioarchitectural parameters within and among tumour cells holds great promise in onconanotomy field, paving the way for a better understanding of tumour tissue 3D organization [7]. ...
... Each organelle or cellular component possesses distinct characteristics, including number, size, shape, texture, or grey level in images. Consequently, their individual segmentations present diverse methodological challenges [7]. Manual segmentation is laborious, necessitating automated methods to comprehend the functioning of a heterogeneous lesion in its entirety. ...
... Additionally, by incorporating a third class representing touching borders, they improved the discriminative capability of the network. Alternative methods utilize a combination of techniques, integrating deep learning, optical flow, and cross-slice linking, to extrapolate segmentations or manual corrections beyond the original plane [4,7,13]. More recently, Isensee et al. [14] presented a self-configuring method for biological and medical image segmentation named nnU-Net. ...
Article
Full-text available
Accurately segmenting and individualizing cells in scanning electron microscopy (SEM) images is a highly promising technique for elucidating tissue architecture in oncology. While current artificial intelligence (AI)-based methods are effective, errors persist, necessitating time-consuming manual corrections, particularly in areas where the quality of cell contours in the image is poor and requires gap filling. This study presents a novel AI-driven approach for refining cell boundary delineation to improve instance-based cell segmentation in SEM images, also reducing the necessity for residual manual correction. A convolutional neural network (CNN) Closing Operator (COp-Net) is introduced to address gaps in cell contours, effectively filling in regions with deficient or absent information. The network takes as input cell contour probability maps with potentially inadequate or missing information and outputs corrected cell contour delineations. The lack of training data was addressed by generating low integrity probability maps using a tailored partial differential equation (PDE). To ensure reproducibility, COp-Net weights and the source code for solving the PDE are publicly available at https://github.com/Florian-40/CellSegm. We showcase the efficacy of our approach in augmenting cell boundary precision using both private SEM images from patient-derived xenograft (PDX) hepatoblastoma tissues and publicly accessible images datasets. The proposed cell contour closing operator exhibits a notable improvement in tested datasets, achieving respectively close to 50% (private data) and 10% (public data) increase in the accurately-delineated cell proportion compared to state-of-the-art methods. Additionally, the need for manual corrections was significantly reduced, therefore facilitating the overall digitalization process. Our results demonstrate a notable enhancement in the accuracy of cell instance segmentation, particularly in highly challenging regions where image quality compromises the integrity of cell boundaries, necessitating gap filling. Therefore, our work should ultimately facilitate the study of tumour tissue bioarchitecture in onconanotomy field.
... This technology enables the visualization of organelles such as nuclei, mitochondria, nucleoli, and lipid droplets and so on, but also the study of the organization of cells into the tissue, an essential step for understanding tumour tissue development mechanisms and drug resistance. Therefore, the analysis of bioarchitectural parameters within and among tumour cells holds great promise in onconanotomy field, paving the way for a better understanding of tumour tissue 3D organization (Senneville et al., 2021). ...
... Each organelle or cellular component possesses distinct characteristics, including number, size, shape, texture, or grey level in images. Consequently, their individual segmentations present diverse methodological challenges (Senneville et al., 2021). Manual segmentation is laborious, necessitating automated methods to comprehend the functioning of a heterogeneous lesion in its entirety. ...
... Additionally, by incorporating a third class representing touching borders, they improved the discriminative capability of the network. Alternative methods utilize a combination of techniques, integrating deep learning, optical flow, and cross-slice linking, to extrapolate segmentations or manual corrections beyond the original plane (Senneville et al., 2021;Machireddy et al., 2023b;Wen et al., 2023). More recently, Isensee et al. (2021) presented a self-configuring method for biological and medical images segmentation named nnU-Net. ...
Preprint
Full-text available
Accurately segmenting and individualizing cells in SEM images is a highly promising technique for elucidating tissue architecture in oncology. While current AI-based methods are effective, errors persist, necessitating time-consuming manual corrections, particularly in areas where the quality of cell contours in the image is poor and requires gap filling. This study presents a novel AI-driven approach for refining cell boundary delineation to improve instance-based cell segmentation in SEM images, also reducing the necessity for residual manual correction. A CNN COp-Net is introduced to address gaps in cell contours, effectively filling in regions with deficient or absent information. The network takes as input cell contour probability maps with potentially inadequate or missing information and outputs corrected cell contour delineations. The lack of training data was addressed by generating low integrity probability maps using a tailored PDE. We showcase the efficacy of our approach in augmenting cell boundary precision using both private SEM images from PDX hepatoblastoma tissues and publicly accessible images datasets. The proposed cell contour closing operator exhibits a notable improvement in tested datasets, achieving respectively close to 50% (private data) and 10% (public data) increase in the accurately-delineated cell proportion compared to state-of-the-art methods. Additionally, the need for manual corrections was significantly reduced, therefore facilitating the overall digitalization process. Our results demonstrate a notable enhancement in the accuracy of cell instance segmentation, particularly in highly challenging regions where image quality compromises the integrity of cell boundaries, necessitating gap filling. Therefore, our work should ultimately facilitate the study of tumour tissue bioarchitecture in onconanotomy field.
... Moreover, metabolite and waste concentrations vary within tumors [3]. Tumor cells interact closely and communicate with the surrounding microenvironment which is comprised of stromal cells and the extracellular matrix (ECM) [3][4][5]. Spheroids are 3D tumor cell structures mimicking some of the bio-architectural characteristics found in tumor tissues. Spheroids with a diameter of 200 μm or more display a metabolic and waste gradient as in tumors, and those with a diameter of 500 μm or more have a hypoxic center [1,6]. ...
Article
Full-text available
Three-dimensional spheroids are more representative of tumors than cell-cultured monolayers. As in tumors, gradients of oxygen, nutrients and wastes are found in spheroid cultures but not in classical cultured monolayers. On the other hand, cell-based assays on the latter are hardly applicable to spheroid cultures. Such is the case for zymogram assays, which are classically used to measure MMP-2 and MMP-9 activities, and for immunoblots to measure the phosphorylation of proteins involved in ligand-induced intracellular signaling in normal and tumor cells. In this study we used two renal cancer cell lines as models, the first derived from a pediatric rhabdoid tumor and the second from an adult clear cell renal cell carcinoma. Using these two cell lines, we successfully developed a simple inexpensive assay to measure MMP-2 and MMP-9 activities in spheroids established in the presence of methylcellulose. After washing, 1 to 5 spheroids were pooled and stimulated with collagen I for 24 h before analysis. MMP-2 and MMP-9 activities were measured in supernatants using a standard but enhanced zymogram assay. Both pro-MMP-9 and MMP-2 activities were detected in spheroids established from both cell lines. In contrast with our previous data using classical cultures monolayers, collagen I stimulation decreased pro-MMP-9 activity without affecting MMP-2 activity. On the other hand, we could not accurately measure AKT intracellular signaling pathways from spheroids stimulated with collagen I. Finally, we adapted our 3D protocol to analyze the MAPK/ERK pathway in kidney tumor cells following induction by EGF. In conclusion, this zymogram assay for analyzing MMP-2 and MMP-9 activities in spheroids paves the way for novel experimentations in tumor biology. Supplementary Information The online version contains supplementary material available at 10.1186/s12896-025-00961-x.
... For example, three-dimensional ultrastructural analysis revealed that distinct compartments of mitochondrial networks within tumors dictated the bioenergetic capacity of non-small cell lung cancer tumors [156]. Strides have also been made in the field of volume electron microscopy, where subcellular components, such as the nucleus and mitochondria, of liver tumors are visualized within their native 3-dimensional organization [157]. Mitochondrial mediated spatial regulation of tumor metabolism has also been elucidated through the lens of fluxomics. ...
Article
Full-text available
Background Bioenergetic remodeling of core energy metabolism is essential to the initiation, survival, and progression of cancer cells through exergonic supply of adenosine triphosphate (ATP) and metabolic intermediates, as well as control of redox homeostasis. Mitochondria are evolutionarily conserved organelles that mediate cell survival by conferring energetic plasticity and adaptive potential. Mitochondrial ATP synthesis is coupled to the oxidation of a variety of substrates generated through diverse metabolic pathways. As such, inhibition of the mitochondrial bioenergetic system by restricting metabolite availability, direct inhibition of the respiratory Complexes, altering organelle structure, or coupling efficiency may restrict carcinogenic potential and cancer progression. Scope of Review Here, we review the role of bioenergetics as the principal conductor of energetic functions and carcinogenesis while highlighting the therapeutic potential of targeting mitochondrial functions. Major Conclusions Mitochondrial bioenergetics significantly contribute to cancer initiation and survival. As a result, therapies designed to limit oxidative efficiency may reduce tumor burden and enhance the efficacy of currently available antineoplastic agents.
... Recently, cells were segmented in EM images of hepatoblastoma patient-derived xenograft tissue. The method involved labeling cells in every 10th image and using optical flow to propagate the contour from these labeled images onto the neighboring images (de Senneville et al., 2021). Propagationbased methods can only propagate the contour of the cells labeled in the ground truth images. ...
Article
Full-text available
Focused ion beam-scanning electron microscopy (FIB-SEM) images can provide a detailed view of the cellular ultrastructure of tumor cells. A deeper understanding of their organization and interactions can shed light on cancer mechanisms and progression. However, the bottleneck in the analysis is the delineation of the cellular structures to enable quantitative measurements and analysis. We mitigated this limitation using deep learning to segment cells and subcellular ultrastructure in 3D FIB-SEM images of tumor biopsies obtained from patients with metastatic breast and pancreatic cancers. The ultrastructures, such as nuclei, nucleoli, mitochondria, endosomes, and lysosomes, are relatively better defined than their surroundings and can be segmented with high accuracy using a neural network trained with sparse manual labels. Cell segmentation, on the other hand, is much more challenging due to the lack of clear boundaries separating cells in the tissue. We adopted a multi-pronged approach combining detection, boundary propagation, and tracking for cell segmentation. Specifically, a neural network was employed to detect the intracellular space; optical flow was used to propagate cell boundaries across the z-stack from the nearest ground truth image in order to facilitate the separation of individual cells; finally, the filopodium-like protrusions were tracked to the main cells by calculating the intersection over union measure for all regions detected in consecutive images along z-stack and connecting regions with maximum overlap. The proposed cell segmentation methodology resulted in an average Dice score of 0.93. For nuclei, nucleoli, and mitochondria, the segmentation achieved Dice scores of 0.99, 0.98, and 0.86, respectively. The segmentation of FIB-SEM images will enable interpretative rendering and provide quantitative image features to be associated with relevant clinical variables.
Article
Tissue microenvironments are extremely complex and heterogeneous. It is challenging to study metabolic interaction between the different cell types in a tissue with the techniques that are currently available. Here we describe a multimodal imaging pipeline that allows cell type identification and nanoscale tracing of stable isotope-labeled compounds. This pipeline extends upon the principles of correlative light, electron and ion microscopy, by combining confocal microscopy reporter or probe-based fluorescence, electron microscopy, stable isotope labeling and nanoscale secondary ion mass spectrometry. We apply this method to murine models of hepatocellular and mammary gland carcinomas to study uptake of glucose derived carbon (13C) and glutamine derived nitrogen (15N) by tumor-associated immune cells. In vivo labeling with fluorescent-tagged antibodies (B220, CD3, CD8a, CD68) in tandem with confocal microscopy allows for the identification of specific cell types (B cells, T cells and macrophages) in the tumor microenvironment. Subsequent image correlation with electron microscopy offers the contrast and resolution to image membranes and organelles. Nanoscale secondary ion mass spectrometry tracks the enrichment of stable isotopes within these intracellular compartments. The whole protocol described here would take ~6 weeks to perform from start to finish. Our pipeline caters to a broad spectrum of applications as it can easily be adapted to trace the uptake and utilization of any stable isotope-labeled nutrient, drug or a probe by defined cellular populations in any tissue in situ.
Article
Structural observations are essential for the advancement of life science. Volume electron microscopy has recently realized remarkable progress in the three-dimensional analyses of biological specimens for elucidating complex ultrastructures in several fields of life science. The advancements in volume electron microscopy technologies have led to improvements, including higher resolution, more stability, and the ability to handle larger volumes. Although human applications of volume electron microscopy remain limited, the reported applications in various organs have already provided previously unrecognized features of human tissues and also novel insights of human diseases. Simultaneously, the application of volume electron microscopy to human studies faces challenges, including ethical and clinical hurdles, costs of data storage and analysis, and efficient and automated imaging methods for larger volume. Solutions including the use of residual clinical specimens and data analysis based on artificial intelligence would address those issues and establish the role of volume electron microscopy in human structural research. Future advancements in volume electron microscopy are anticipated to lead to transformative discoveries in basic research and clinical practice, deepening our understanding of human health and diseases for better diagnostic and therapeutic strategies.
Article
Full-text available
Advances in cryogenic electron microscopy (cryoEM) single particle analysis have revolutionized structural biology by facilitating the in vitro determination of atomic- and near-atomic-resolution structures for fully hydrated macromolecular complexes exhibiting compositional and conformational heterogeneity across a wide range of sizes. Cryogenic electron tomography (cryoET) and subtomogram averaging are rapidly progressing toward delivering similar insights for macromolecular complexes in situ , without requiring tags or harsh biochemical purification. Furthermore, cryoET enables the visualization of cellular and tissue phenotypes directly at molecular, nanometric resolution without chemical fixation or staining artifacts. This forward-looking review covers recent developments in cryoEM/ET and related technologies such as cryogenic focused ion beam milling scanning electron microscopy and correlative light microscopy, increasingly enhanced and supported by artificial intelligence algorithms. Their potential application to emerging concepts is discussed, primarily the prospect of complementing medical histopathology analysis. Machine learning solutions are poised to address current challenges posed by “big data” in cryoET of tissues, cells, and macromolecules, offering the promise of enabling novel, quantitative insights into disease processes, which may translate into the clinic and lead to improved diagnostics and targeted therapeutics.
Article
Full-text available
Brain structure segmentation on magnetic resonance (MR) images is important for various clinical applications. It has been automatically performed by using fully convolutional networks. However, it suffers from the class imbalance problem. To address this problem, we investigated how loss weighting strategies work for brain structure segmentation tasks with different class imbalance situations on MR images. In this study, we adopted segmentation tasks of the cerebrum, cerebellum, brainstem, and blood vessels from MR cisternography and angiography images as the target segmentation tasks. We used a U-net architecture with cross-entropy and Dice loss functions as a baseline and evaluated the effect of the following loss weighting strategies: inverse frequency weighting, median inverse frequency weighting, focal weighting, distance map-based weighting, and distance penalty term-based weighting. In the experiments, the Dice loss function with focal weighting showed the best performance and had a high average Dice score of 92.8% in the binary-class segmentation tasks, while the cross-entropy loss functions with distance map-based weighting achieved the Dice score of up to 93.1% in the multi-class segmentation tasks. The results suggested that the distance map-based and the focal weightings could boost the performance of cross-entropy and Dice loss functions in class imbalanced segmentation tasks, respectively.
Article
Full-text available
The notion of tumor microenvironment (TME) has been brought to the forefront of recent scientific literature on cancer. However, there is no consensus on how to define and spatially delineate the TME. We propose that the time is ripe to go beyond an all-encompassing list of the components of the TME, and to construct a multilayered view of cancer. We distinguish six layers of environmental interactions with the tumor and show that they are associated with distinct mechanisms, and ultimately with distinct therapeutic approaches.
Article
Full-text available
Recent developments in serial-section electron microscopy allow the efficient generation of very large image data sets but analyzing such data poses challenges for software tools. Here we introduce Volume Annotation and Segmentation Tool (VAST), a freely available utility program for generating and editing annotations and segmentations of large volumetric image (voxel) data sets. It provides a simple yet powerful user interface for real-time exploration and analysis of large data sets even in the Petabyte range.
Article
Full-text available
Efficient and accurate semantic segmentation is the key technique for automatic remote sensing image analysis. While there have been many segmentation methods based on traditional hand-craft feature extractors, it is still challenging to process high-resolution and large-scale remote sensing images. In this work, a novel patch-wise semantic segmentation method with a new training strategy based on fully convolutional networks is presented to segment common land resources. First, to handle the high-resolution image, the images are split as local patches and then a patch-wise network is built. Second, training data is preprocessed in several ways to meet the specific characteristics of remote sensing images, i.e., color imbalance, object rotation variations and lens distortion. Third, a multi-scale training strategy is developed to solve the severe scale variation problem. In addition, the impact of conditional random field (CRF) is studied to improve the precision. The proposed method was evaluated on a dataset collected from a capital city in West China with the Gaofen-2 satellite. The dataset contains ten common land resources (Grassland, Road, etc.). The experimental results show that the proposed algorithm achieves 54.96% in terms of mean intersection over union (MIoU) and outperforms other state-of-the-art methods in remote sensing image segmentation.
Article
Full-text available
Hepatoblastoma (HBL) is a pediatric liver cancer with defined molecular alterations driving its progression. Here, we describe an animal model for HBL on the chick chorioallantoic membrane (CAM), which recapitulates relevant features of HBL in patients. Expression of classic tumor-associated proteins such as β-catenin, EpCAM and CK19 was maintained in acini-like organized tumors on CAM, as was synthesis of AFP, a tumor marker used for monitoring patient response. RNA sequencing revealed an unexpected molecular evolution of HBL cells on the CAM, with significant deregulation of more than 6,000 genes including more than half of all HOX genes. Bioinformatic analysis distinguish between tumor cell-expressed genes and chick genes, thereby shedding new light on the complex interactions taking place during HBL progression. Importantly, human tumor suppressive ribosomal genes were downregulated after implantation, whereas mitochondrial genes encoding for anti-apoptotic peptides were strongly induced in vivo. Meprin-1α expression was increased during evolution of CAM tumors and confirmed by immunohistochemistry. Cisplatin, a commonly used chemotherapeutic agent for HBL, showed significant anti-tumoral effects. Our results broaden the understanding of the molecular adaptation process of human cancer cells to the microenvironment and might help to elaborate novel therapeutic concepts for the treatment of this pediatric liver tumor.
Article
Full-text available
Surgery and cisplatin-based treatment of hepatoblastoma (HB) currently guarantee the survival of 70-80% of patients. However, some important challenges remain in diagnosing high risk tumors and identifying relevant targetable pathways offering new therapeutic avenues. Previously, two molecular subclasses of hepatoblastoma tumors have been described, namely C1 and C2; C2 being the subgroup with the poorest prognosis, a more advanced tumor stage and the worst overall survival rate. An associated 16-gene signature to discriminate the two tumoral subgroups was proposed but it has not been transferred into clinical routine. To address these issues we performed RNA sequencing of 25 tumors and matched normal liver samples from patients. The transcript profiling separated HB into three distinct subgroups named C1, C2A and C2B, identifiable by a concise four-gene signature: HSD17B6, ITGA6, TOP2A and VIM, with TOP2A being characteristic for the proliferative C2A tumors. Differential expression of these genes was confirmed by RT-qPCR on an expanded cohort and by immunohistochemistry. We also revealed significant overexpression of genes involved in Fanconi Anemia (FA) pathway in the C2A subgroup. We then investigated the ability of several described FA inhibitors to block growth of HB cells in vitro and in vivo. We demonstrated that bortezomib, an FDA-approved proteasome inhibitor, strongly impairs the proliferation and survival of HB cell lines in vitro, blocks FA pathway associated double-strand DNA repair and significantly impedes HB growth in vivo. In conclusion, the highly proliferating C2A subtype is characterized by TOP2A gene up-regulation and FA pathway activation and HB therapeutic arsenal could include Bortezomib for the treatment of patients with the most aggressive tumors. Published Poster award awarded by the International Society of Paediatric Oncology (50th SIOP Congress)
Chapter
Recent developments in large format electron microscopy have enabled generation of images that provide detailed ultrastructural information on normal and diseased cells and tissues. Analyses of these images increase our understanding of cellular organization and interactions and disease-related changes therein. In this manuscript, we describe a workflow for two-dimensional (2D) and three-dimensional (3D) imaging, including both optical and scanning electron microscopy (SEM) methods, that allow pathologists and cancer biology researchers to identify areas of interest from human cancer biopsies. The protocols and mounting strategies described in this workflow are compatible with 2D large format EM mapping, 3D focused ion beam-SEM and serial block face-SEM. The flexibility to use diverse imaging technologies available at most academic institutions makes this workflow useful and applicable for most life science samples. Volumetric analysis of the biopsies studied here revealed morphological, organizational and ultrastructural aspects of the tumor cells and surrounding environment that cannot be revealed by conventional 2D EM imaging. Our results indicate that although 2D EM is still an important tool in many areas of diagnostic pathology, 3D images of ultrastructural relationships between both normal and cancerous cells, in combination with their extracellular matrix, enables cancer researchers and pathologists to better understand the progression of the disease and identify potential therapeutic targets.
Article
We present ilastik, an easy-to-use interactive tool that brings machine-learning-based (bio)image analysis to end users without substantial computational expertise. It contains pre-defined workflows for image segmentation, object classification, counting and tracking. Users adapt the workflows to the problem at hand by interactively providing sparse training annotations for a nonlinear classifier. ilastik can process data in up to five dimensions (3D, time and number of channels). Its computational back end runs operations on-demand wherever possible, allowing for interactive prediction on data larger than RAM. Once the classifiers are trained, ilastik workflows can be applied to new data from the command line without further user interaction. We describe all ilastik workflows in detail, including three case studies and a discussion on the expected performance. ilastik is an user-friendly interactive tool for machine-learning-based image segmentation, object classification, counting and tracking.
Article
Detailed whole brain segmentation is an essential quantitative technique in medical image analysis, which provides a non-invasive way of measuring brain regions from a clinical acquired structural magnetic resonance imaging (MRI). Recently, deep convolution neural network (CNN) has been applied to whole brain segmentation. However, restricted by current GPU memory, 2D based methods, downsampling based 3D CNN methods, and patch-based high-resolution 3D CNN methods have been the de facto standard solutions. 3D patch-based high resolution methods typically yield superior performance among CNN approaches on detailed whole brain segmentation (>100 labels), however, whose performance are still commonly inferior compared with state-of-the-art multi-atlas segmentation methods (MAS) due to the following challenges: (1) a single network is typically used to learn both spatial and contextual information for the patches, (2) limited manually traced whole brain volumes are available (typically less than 50) for training a network. In this work, we propose the spatially localized atlas network tiles (SLANT) method to distribute multiple independent 3D fully convolutional networks (FCN) for high-resolution whole brain segmentation. To address the first challenge, multiple spatially distributed networks were used in the SLANT method, in which each network learned contextual information for a fixed spatial location. To address the second challenge, auxiliary labels on 5111 initially unlabeled scans were created by multi-atlas segmentation for training. Since the method integrated multiple traditional medical image processing methods with deep learning, we developed a containerized pipeline to deploy the end-to-end solution. From the results, the proposed method achieved superior performance compared with multi-atlas segmentation methods, while reducing the computational time from >30 h to 15 min. The method has been made available in open source (https://github.com/MASILab/SLANTbrainSeg).
Article
Cells control their size through an intricate balance of cell growth, cell division, and cell death. Extensive work on unicellular model organisms revealed that cell-size-dependent cell cycle progression accounts for major aspects of cell size regulation and provided insights into the underlying molecular mechanisms. Nevertheless, elaborate live-cell imaging approaches still reveal new phenomenological observations that challenge our simplified models of size regulation and raise the question of what determines optimal cell size. Here, I aim to give a conceptual overview of the many processes contributing to cell size regulation and summarize recent developments in the field.