Macrophage diversity in cancer revisited in the era of single-cell omics
Ruoyu Ma,
No information about this author
Annabel Black,
No information about this author
Bin‐Zhi Qian
No information about this author
et al.
Trends in Immunology,
Journal Year:
2022,
Volume and Issue:
43(7), P. 546 - 563
Published: June 9, 2022
TAMs
have
diverse
functions
in
cancer,
reflecting
the
heterogenous
nature
of
these
immune
cells.
Here,
we
propose
a
new
nomenclature
to
identify
TAM
subsets.Recent
single
cell
multi-omics
technologies,
which
allow
clustering
subsets
an
unbiased
manner,
significantly
advanced
our
understanding
molecular
diversity
mice
and
humans.Novel
mechanisms
potential
therapeutic
targets
been
identified
that
might
regulate
tumor-promoting
function
different
subsets.TAM
opens
promising
opportunities
for
envisaging
putative
cancer
treatments.
Tumor-associated
macrophages
(TAMs)
multiple
potent
and,
thus,
represent
important
targets.
These
highlight
TAMs.
Recent
omics
technologies
However,
unifying
annotation
their
signatures
is
lacking.
review
recent
major
studies
transcriptome,
epigenome,
metabolome,
spatial
with
specific
focus
on
We
also
consensus
model
present
avenues
future
research.
one
most
abundant
types
tumors
[1.Cassetta
L.
Pollard
J.W.
Targeting
macrophages:
approaches
cancer.Nat.
Rev.
Drug
Discov.
2018;
17:
887-904Crossref
PubMed
Scopus
(650)
Google
Scholar].
Since
initial
decade
ago
[2.Qian
B.Z.
Macrophage
enhances
tumor
progression
metastasis.Cell.
2010;
141:
39-51Abstract
Full
Text
PDF
(3151)
Scholar],
functional
now
widely
appreciated,
many
seminal
field
[3.Yang
M.
et
al.Diverse
microenvironments.Cancer
Res.
78:
5492-5503Crossref
(202)
Scholar,
4.DeNardo
D.G.
Ruffell
B.
Macrophages
as
regulators
tumour
immunity
immunotherapy.Nat.
Immunol.
2019;
19:
369-382Crossref
(643)
5.Lopez-Yrigoyen
al.Macrophage
targeting
cancer.Ann.
N.
Y.
Acad.
Sci.
2021;
1499:
18-41Crossref
(25)
This
array
includes
promotion
growth,
lineage
plasticity,
invasion,
remodeling
extracellular
matrix,
crosstalk
endothelial,
mesenchymal
stromal
cells,
other
cells;
effects
can
result
progression,
metastasis
(see
Glossary),
therapy
resistance
[6.Mantovani
A.
al.Tumour-associated
treatment
oncology.Nat.
Clin.
Oncol.
2017;
14:
399-416Crossref
(1675)
Scholar,7.Guc
E.
Redefining
macrophage
neutrophil
biology
metastatic
cascade.Immunity.
54:
885-902Abstract
(13)
With
wide
application
years
seen
explosion
data
illustrating
cellular
heterogeneity
resulting
unprecedented
amount
information
TAMs,
regardless
main
studies.
Links
between
are
emerging.
terminology
lacking,
making
direct
comparisons
full
utilization
sets
difficult.
In
this
review,
summarize
human
data;
include
traditional
nomenclatures,
at
levels
single-cell
transcriptomic,
epigenomic,
metabolic
multi-omics,
opportunities,
directions.
subsets.
hope
will
serve
starting
point
help
build
complete
picture
dynamic
interactions
tumor,
well
microenvironment
(TME).
A
used
describe
has
now-obsolete
M1/M2
model,
proposed
~20
ago;
it
separated
into
two
distinct
arms:
M1
or
'classically'
activated;
M2
'alternatively'
activated,
largely
based
vitro
stimulating
type
1
2
cytokines
[8.Mills
C.D.
al.M-1/M-2
Th1/Th2
paradigm.J.
2000;
164:
6166-6173Crossref
The
newer
term
'M1-like'
phenotype
typically
described
proinflammatory
induced
by
Toll-like
receptor
(TLR)
ligands
cytokines,
namely
IFN-γ
TNF-α.
Conversely,
'M2-like'
having
anti-inflammatory
characteristics,
being
activated
interleukin
(IL)-4
IL-13,
producing
TGF-β
profibrotic
factors.
nomenclature,
albeit
used,
remains
oversimplified
[9.Martinez
F.O.
Gordon
S.
paradigm
activation:
time
reassessment.F1000Prime
Rep.
2014;
6:
13Crossref
(2673)
Scholar,10.Nahrendorf
Swirski
F.K.
Abandoning
network
function.Circ.
2016;
119:
414-417Crossref
(195)
Indeed,
significant
morphology,
function,
surface
marker
expression
observed
resident-tissue
(RTMs)
from
organs
[11.Bleriot
C.
al.Determinants
resident
tissue
identity
function.Immunity.
2020;
52:
957-970Abstract
(94)
Scholar];
moreover,
co-expression
both
gene
almost
all
[12.Mulder
K.
al.Cross-tissue
landscape
monocytes
health
disease.Immunity.
1883-1900Abstract
Therefore,
spectrum
polarization
relates
represents
more
sensible
approach
describing
[10.Nahrendorf
Scholar,13.Mosser
D.M.
Edwards
J.P.
Exploring
activation.Nat.
2008;
8:
958-969Crossref
(5864)
normal
homeostasis,
tightly
regulated
niche-like
local
environment,
recently
[14.Guilliams
al.Establishment
maintenance
niche.Immunity.
434-451Abstract
(138)
Another
layer
derives
origin.
Using
lineage-tracing
mice,
illustrated
mouse
RTMs
derived
early
erythromyeloid
progenitors
formed
either
yolk
sac
fetal
liver
[15.Geissmann
F.
al.Blood
consist
principal
migratory
properties.Immunity.
2003;
71-82Abstract
(2514)
Scholar,16.Gomez
Perdiguero
al.Tissue-resident
originate
yolk-sac-derived
erythro-myeloid
progenitors.Nature.
2015;
518:
547-551Crossref
(1236)
Additionally,
adult
may
derive
circulating
monocytic
precursors
(monocytes)
bone
marrow
[17.Cox
al.Origins,
biology,
diseases
macrophages.Annu.
39:
313-344Crossref
(1)
monocyte
contribution
varies
among
organs.
For
example,
steady
state,
microglia
central
nervous
system
(CNS)
solely
[18.Hoeffel
G.
al.C-Myb(+)
progenitor-derived
give
rise
tissue-resident
macrophages.Immunity.
42:
665-678Abstract
(611)
while
dermal
embryonic
origin
[19.Kolter
J.
al.A
subset
skin
contributes
surveillance
regeneration
nerves.Immunity.
50:
1482-1497Abstract
(69)
appreciated
repeatedly
reviewed
[20.Pathria
P.
al.Targeting
tumor-associated
cancer.Trends
40:
310-327Abstract
(382)
Scholar,21.Guerriero
J.L.
Macrophages:
road
less
traveled,
changing
anticancer
therapy.Trends
Mol.
Med.
24:
472-489Abstract
(143)
Similar
counterparts
not
only
its
ontogeny,
but
cues,
including
type,
organ,
subanatomic
Identifying
basis
over
past
[5.Lopez-Yrigoyen
advancements
unveiling
multidimensional
complexity
manner.
research,
oncology
eventually
fully
understand
cells
hopefully
use
improve
precision
diagnosis
therapy.
Single
RNA
sequencing
(scRNA-seq)
technology
revolutionized
providing
in-depth
transcriptome
level
[22.Giladi
al.Single-cell
characterization
haematopoietic
trajectories
homeostasis
perturbed
haematopoiesis.Nat.
Cell
Biol.
20:
836-846Crossref
(139)
substantial
advances
available
experimental
techniques
bioinformatics
pipelines
years,
scRNA-seq
investigate
[23.Lawson
D.A.
al.Tumour
resolution.Nat.
1349-1360Crossref
(230)
Scholar,24.Ren
X.
al.Insights
gained
analysis
microenvironment.Annu.
583-609Crossref
(15)
transcriptomic
remain
Two
large-scale
pan-cancer
provided
valuable
regarding
diversity.
One
study
analyzed
myeloid
380
samples
across
15
210
patients
through
combination
newly
collected
eight
published
[25.Cheng
transcriptional
atlas
infiltrating
cells.Cell.
184:
792-809Abstract
(111)
Comparison
consistent
presence
CD14+
CD16+
tumor-infiltrating
(TIMs),
LYVE1+
interstitial
non-cancer
tissues,
seven
clusters:
INHBA+
C1QC+
ISG15+
LNRP3+
SPP1+
compiled
mononuclear
phagocytes
(MNPs)
isolated
41
13
types,
six
common
universe,
termed
MNP-VERSE.
Monocyte
clusters
were
then
extracted
reintegrated
generate
MoMac-VERSEi.
regulatory
inference
(SCENIC)
[26.Aibar
al.SCENIC:
clustering.Nat.
Methods.
1083-1086Crossref
(1003)
authors
classical
monocytes,
nonclassical
five
(HES1
TAM,
C1Qhi
TREM2
IL4I1
proliferating
TAMs)
Although
nomenclatures
studies,
others,
pattern
transcriptomics
By
reviewing
journals,
found
preserved
(Table
1).
Based
signature
genes,
enriched
pathways,
predicated
naming
interferon-primed
(IFN-TAMs),
(Reg-TAMs),
inflammatory
cytokine-enriched
(Inflam-TAMs),
lipid-associated
(LA-TAMs),
pro-angiogenic
(Angio-TAMs),
RTM-like
(RTM-TAMs),
(Prolif-TAMs)
Figure
1,
Key
figure).
Furthermore,
three
TIMs
Box
1).Table
1Mouse
various
TMEsaBlack
font:
genes
clusters;
blue
protein
markers
Underline:
CITE-seq;
Bold:
key
reported
than
paper.,
bAbbreviations:
BRCA,
breast
cancer;
CAF,
cancer-associated
macrophage;
CITE-seq,
indexing
transcriptomes
epitopes
sequencing;
CRC,
colorectal
CyTOF,
Mass
cytometry
flight;
ECM,
matrix;
ESCA,
esophageal
carcinoma;
GC,
gastric
HCC,
hepatocellular
HNC,
head
neck
i.v.,
intravenous;
IF,
immunofluorescent
staining;
INs-seq,
intracellular
staining
LCM,
laser
capture
microdissection;
LYM,
lymphoma;
MEL,
melanoma;
Mets,
metastasis;
mIHC,
multiplex
immunochemistry
MMY,
myeloma;
N/A,
available;
NPC,
nasopharyngeal
NSCLC,
nonsmall
lung
OS,
osteosarcoma;
OVC,
ovarian
PDAC,
pancreatic
ductal
adenocarcinoma;
PRAC,
prostate
RCC,
renal
Reg-TAMs,
TAMs;
SARC,
sarcoma;
sc-MS,
mass
spectrometry;
SEPN,
spinal
ependymomas;
SKC,
ST,
transcriptomics;
s.c.,
subcutaneous;
macrophages;
THCA,
thyroid
UCEC,
uterine
corpus
endometrial
carcinoma.AnnotationSpeciesSignatureTFCancer
typeFunction/enriched
pathwayAssayRefsIFN-TAMsHumanCASP1,
CASP4,
CCL2/3/4/7/8,
CD274hi,
CD40,
CXCL2/3/9/10/11,
IDO1,
IFI6,
IFIT1/2/3,
IFITM1/3,
IRF1,
IRF7,
ISG15,
LAMP3,
PDCD1LG2hi,
TNFSF10,
C1QA/C,
CD38,
IL4I1,
IFI44LSTAT1
IRF1/7BRCACRCCRC
metsGBMHCCHNCLYMMELMMYNPCNSCLCOSPDACSEPNTHCAUCECApoptosis
regulatorsEnhance
proliferationInflammatory
responsesPromote
Treg
entry
tumorT
exhaustionImmunosuppressionColocalization
exhausted
T
(ST,
IF)Decreased
antigen
presentation
(CyTOF)Suppressed
activation
(in
vitro)IFN-α/γ-IFN
response
signature;
IL2/STAT5;
IL6/JAK/STAT3scRNA-seqCITE-seqmIHCSTNanoString
GeoMx[12.Mulder
Scholar,29.Gubin
M.M.
al.High-dimensional
delineates
lymphoid
compartment
during
successful
immune-checkpoint
therapy.Cell.
175:
1014-1030Abstract
(165)
Scholar,32.Zavidij
O.
reveals
compromised
precursor
stages
myeloma.Nat.
Cancer.
1:
493-506Crossref
33.Zhou
intratumoral
immunosuppressive
osteosarcoma.Nat.
Commun.
11:
6322Crossref
(74)
34.Zhang
Q.
al.Interrogation
microenvironmental
ependymomas
dual
macrophages.Nat.
12:
6867Crossref
(0)
Scholar,45.Wu
al.Spatiotemporal
level.Cancer
134-153Crossref
(10)
Scholar,52.Pombo
Antunes
A.R.
profiling
glioblastoma
species
disease
stage
competition
specialization.Nat.
Neurosci.
595-610Crossref
(78)
Scholar,\81.Wu
S.Z.
spatially
resolved
cancers.Nat.
Genet.
53:
1334-1347Crossref
(47)
Scholar,83.Pelka
al.Spatially
organized
multicellular
hubs
cancer.Cell.
4734-4752Abstract
(29)
Scholar]CD14+,
CD11b+,
CD68+,
PD-L1hi,
PD-L2hi,
CD80hi,
CD86hi,
MHCIIhi,
CD86+,
MRC1–,
SIGLEC1–,
HLA-DRlo,
CD314+,
CD107a+,
CD86,
TLR4,
CD44
(CITE-seq)MouseCcl2/7/8,
Cd274,
Cxcl9/10/11,
Ifit1/2/3,
Ifit3,
Ifitm1/3,
Il7r,
Isg15,
Nos2,
Rsad2,
Tnfsf10,
Stat1N/ACT26
s.c.
CRCCT26
intrasplenic
mets
modelT3
SARC
(s.c.)Orthotopic
GL261
GBMIFN
signaturescRNA-seqCITE-seqmIHC[29.Gubin
Scholar]Inflam-TAMsHumanCCL2/3/4/5/20,
CCL3L1,
CCL3L3,
CCL4L2,
CCL4L4,
CXCL1/2/3/5/8,
G0S2,
IL1B,
IL1RN,
IL6,
INHBA,
KLF2/6,
NEDD9,
PMAIP1,
S100A8/A9,
SPP1EGR3
IKZF1
NFKB1
NFE2L2
RELCRCCRC
metsOSSEPNGCRecruiting
regulating
cellsCNS
inflammation-associated
chemokinesPromotes
inflammationNeutrophil
recruitment
lumenT
interaction
(IHC)TNF
signaling;
WNTImmune
check
pointsscRNA-seqmIHCNanoString
GeoMx[31.Che
L.-H.
metastases
reprogramming
preoperative
chemotherapy.Cell
Discovery.
7:
80Crossref
(4)
Scholar,33.Zhou
Scholar,34.Zhang
Scholar,42.Sathe
genomic
microenvironment.Clin.
Cancer
26:
2640-2653Crossref
(66)
43.Zhang
al.Dissecting
underlying
premalignant
lesions
cancer.Cell
27:
1934-1947Abstract
(104)
44.Yin
H.
map
development
using
sequencing.Front.
12728169Crossref
45.Wu
Scholar]MouseCxcl1/2/3/5/8,
Ccl20,
Ccl3l1,
Il1rn,
Il1b,
G0s2,
Inhba,
Spp1N/ACT26
CRC
CT26
modelChemokine
productionImmunosuppressionscRNA-seq[45.Wu
Scholar]LA-TAMsHumanACP5,
AOPE,
APOC1,
ATF1,
C1QA/B/C,
CCL18,
CD163,
CD36,
CD63,
CHI3L1,
CTSB/D/L,
F13A1,
FABP5,
FOLR2,
GPNMB,
IRF3,
LGALS3,
LIPA,
LPL,
MACRO,
MerTK,
MMP7/9/12,
MRC1,
NR1H3,
NRF1,
NUPR1,
PLA2G7,
RNASE1,
SPARC,
SPP1,
TFDP2,
TREM2,
ZEB1FOS/JUN
HIF1A
MAF/MAFB
NR1H3
TCF4
TFECBRCACRCCRC
metsGBMGCHCCHNCNPCNSCLCOSPDACPhagocytosisPromotion
EMTComplement
activationECM
degradationAntigen
processing
pathwaysATP
biosynthetic
processesCanonical
M2-like
pathwaysFatty
acid
metabolismImmunosuppressionInflammationIron
ion
signalingscRNA-seqSMART-seq2CITE-seqmIHCST[12.Mulder
Scholar,27.Zilionis
R.
cancers
conserved
populations
individuals
species.Immunity.
1317-1334Abstract
(424)
Scholar,28.Yang
non-small
differences
sexes.Front.
12756722Google
Scholar,30.Zhang
analyses
inform
myeloid-targeted
therapies
colon
181:
442-459Abstract
(246)
Scholar,31.Che
Scholar,50.Chen
Y.P.
subtypes
associated
prognosis
carcinoma.Cell
30:
1024-1042Crossref
(71)
Scholar,81.Wu
Scholar]CD9+,
CD80+,
MAF,
CD163lo/-,
CD206+/lo,
CD71+,
CD72+,
CD73,
ICOSL,
CD40LG,
Thy-1
(CITE-seq)MouseAcp5,
Apoc1,
Apoe,
C1qa/B/C,
Ccl18,
Ccl8,
Cd163,
Cd206,
Cd36,
Cd63,
Ctsb/d/l,
Cxcl9,
Fabp5,
Folr2,
Gpnmb,
Lgals3,
Macro,
Mrc1,
Trem2MAFCT26
Orthotopic
GBM
7940b
orthotopic
iKras
p53
PDAC
metsPhagocytosisAntigen
presentationFatty
metabolismComplement
activationscRNA-seqCITE-seqmIHC[45.Wu
Scholar,46.Kemp
S.B.
al.Pancreatic
marked
complement-high
blood
tumor–associated
macrophages.Life
Alliance.
4e202000935Crossref
Scholar]Angio-TAMsHumanADAM8,
AREG,
BNIP3,
CCL2/4/20,
CD300E,
CD44,
CD55,
CEBPB,
CLEC5A,
CTSB,
EREG,
FCN1,
FLT1,
FN1,
HES1,
IL8,
MIF,
OLR1,
PPARG,
S100A8/9/12,
SERPINB2,
SLC2A1,
SPIC,
THBS1,
TIMP1,
VCAN,
VEGFABACH1
CEBPB
FOSL2
HIFA
KLF5
MAF
RUNX1
SPIC
TEAD1
ZEB2BRCACRCCRCCRC
metsESCAGBMGCHCCMELNPCNPCNSCLCOVCPDACPDAC
metsRCCSEPNTHCAUCECAngiogenesisCAF
interactionECM
proteolysis;
ECM
interactionPromotion
EMTHIF
pathway;
NF-kB
Notch
VEGF
signalingJuxtaposed
PLVAP+/DLL4+
endothelial
(IF)scRNA-seqSMART-seq2CITE-seqNanoString
GeoMx[25.Cheng
Scholar,41.Sharma
al.Onco-fetal
drives
carcinoma.Cell.
183:
377-394Abstract
(103)
Scholar,49.Raghavan
al.Microenvironment
drug
6119-6137Abstract
Scholar,67.Zhao
revealed
promoted
progression.J.
Transl.
454Crossref
Scholar]CD52hi,
CD163hi,
CD206hi,
CXCR4+,
CD354+,
FOSL2,
VEGFAMouseArg1,
Adam8,
Bnip3,
Mif,
Slc2a1N/AOrthotopic
modelHIF
signalingAngiogenesisscRNA-seqCITE-seq[52.Pombo
Scholar]Reg-TAMsHumanCCL2,
CD274,
CD80,
CHIT1,
CX3CR1,
HLA-A/C,
HLA-DQA1/B1,
HLA-DRA/B1/B5,
ICOSLG,
IL-10,
ITGA4,
LGALS9,
MAC
Language: Английский
Applications of single-cell sequencing in cancer research: progress and perspectives
Yalan Lei,
No information about this author
Rong Tang,
No information about this author
Jin Xu
No information about this author
et al.
Journal of Hematology & Oncology,
Journal Year:
2021,
Volume and Issue:
14(1)
Published: June 9, 2021
Single-cell
sequencing,
including
genomics,
transcriptomics,
epigenomics,
proteomics
and
metabolomics
is
a
powerful
tool
to
decipher
the
cellular
molecular
landscape
at
single-cell
resolution,
unlike
bulk
which
provides
averaged
data.
The
use
of
sequencing
in
cancer
research
has
revolutionized
our
understanding
biological
characteristics
dynamics
within
lesions.
In
this
review,
we
summarize
emerging
technologies
recent
progress
obtained
by
information
related
landscapes
malignant
cells
immune
cells,
tumor
heterogeneity,
circulating
underlying
mechanisms
behaviors.
Overall,
prospects
facilitating
diagnosis,
targeted
therapy
prognostic
prediction
among
spectrum
tumors
are
bright.
near
future,
advances
will
undoubtedly
improve
highlight
potential
precise
therapeutic
targets
for
patients.
Language: Английский
Cancer-associated fibroblasts in the single-cell era
Nature Cancer,
Journal Year:
2022,
Volume and Issue:
3(7), P. 793 - 807
Published: July 26, 2022
Language: Английский
Single‐Cell, Single‐Nucleus, and Spatial RNA Sequencing of the Human Liver Identifies Cholangiocyte and Mesenchymal Heterogeneity
Hepatology Communications,
Journal Year:
2021,
Volume and Issue:
6(4), P. 821 - 840
Published: Nov. 18, 2021
The
critical
functions
of
the
human
liver
are
coordinated
through
interactions
hepatic
parenchymal
and
non-parenchymal
cells.
Recent
advances
in
single-cell
transcriptional
approaches
have
enabled
an
examination
with
unprecedented
resolution.
However,
dissociation-related
cell
perturbation
can
limit
ability
to
fully
capture
liver's
fraction,
which
limits
comprehensively
profile
this
organ.
Here,
we
report
landscape
73,295
cells
from
using
matched
RNA
sequencing
(scRNA-seq)
single-nucleus
(snRNA-seq).
addition
snRNA-seq
characterization
interzonal
hepatocytes
at
a
resolution,
revealed
presence
rare
subtypes
mesenchymal
cells,
facilitated
detection
cholangiocyte
progenitors
that
had
only
been
observed
during
vitro
differentiation
experiments.
T
B
lymphocytes
natural
killer
were
distinguishable
scRNA-seq,
highlighting
importance
applying
both
technologies
obtain
complete
map
tissue-resident
types.
We
validated
distinct
spatial
distribution
hepatocyte,
cholangiocyte,
populations
by
independent
transcriptomics
data
set
immunohistochemistry.
Conclusion:
Our
study
provides
systematic
comparison
transcriptomes
captured
scRNA-seq
delivers
high-resolution
healthy
liver.
Language: Английский
Hepatic stellate cells in physiology and pathology
The Journal of Physiology,
Journal Year:
2022,
Volume and Issue:
600(8), P. 1825 - 1837
Published: March 21, 2022
Hepatic
stellate
cells
(HSCs)
comprise
a
minor
cell
population
in
the
liver
but
serve
numerous
critical
functions
normal
and
response
to
injury.
HSCs
are
primarily
known
for
their
activation
upon
injury
producing
collagen-rich
extracellular
matrix
fibrosis.
In
absence
of
injury,
reside
quiescent
state,
which
main
function
appears
be
storage
retinoids
or
vitamin
A-containing
metabolites.
Less
appreciated
include
amplifying
hepatic
inflammatory
expressing
growth
factors
that
development
both
initiation
termination
regeneration.
Recent
single-cell
RNA
sequencing
studies
have
corroborated
earlier
indictaing
HSC
involves
diverse
array
phenotypic
alterations
identified
unique
populations.
This
review
serves
highlight
these
many
HSCs,
briefly
describe
recent
genetic
tools
will
help
thoroughly
investigate
role
physiology
pathology.
Language: Английский
A single cell atlas of the human liver tumor microenvironment
Hassan Massalha,
No information about this author
Keren Bahar Halpern,
No information about this author
Samir Abu‐Gazala
No information about this author
et al.
Molecular Systems Biology,
Journal Year:
2020,
Volume and Issue:
16(12)
Published: Dec. 1, 2020
Article17
December
2020Open
Access
Transparent
process
A
single
cell
atlas
of
the
human
liver
tumor
microenvironment
Hassan
Massalha
orcid.org/0000-0002-9923-6878
Department
Molecular
Cell
Biology,
Weizmann
Institute
Science,
Rehovot,
Israel
Search
for
more
papers
by
this
author
Keren
Bahar
Halpern
Samir
Abu-Gazala
General
Surgery,
Hadassah
Hebrew
University
Medical
Center,
Jerusalem,
Transplant
Division,
Hospital
Pennsylvania,
Philadelphia,
PA,
USA
Tamar
Jana
Efi
E
Massasa
Andreas
Moor
orcid.org/0000-0001-8715-8449
Biosystems
Science
and
Engineering,
ETH
Zürich,
Basel,
Switzerland
Lisa
Buchauer
orcid.org/0000-0002-4722-8390
Milena
Rozenberg
Eli
Pikarsky
The
Lautenberg
Center
Immunology,
Research
Israel-Canada,
School,
Ido
Amit
Gideon
Zamir
Shalev
Itzkovitz
Corresponding
Author
[email
protected]
orcid.org/0000-0003-0685-2522
Information
Massalha1,
Halpern1,
Abu-Gazala2,3,
Jana1,
Massasa1,
Moor4,
Buchauer1,
Rozenberg1,
Pikarsky5,
Amit6,
Zamir2
*,1
1Department
2Department
3Transplant
4Department
5The
6Department
*Corresponding
author.
Tel:
+972
89343104;
E-mail:
Systems
Biology
(2020)16:e9682https://doi.org/10.15252/msb.20209682
PDFDownload
PDF
article
text
main
figures.
Peer
ReviewDownload
a
summary
editorial
decision
including
letters,
reviewer
comments
responses
to
feedback.
ToolsAdd
favoritesDownload
CitationsTrack
CitationsPermissions
ShareFacebookTwitterLinked
InMendeleyWechatReddit
Figures
&
Info
Abstract
Malignant
growth
is
fueled
interactions
between
cells
stromal
composing
microenvironment.
major
site
tumors
metastases,
but
molecular
identities
intercellular
different
types
have
not
been
resolved
in
these
pathologies.
Here,
we
apply
RNA-sequencing
spatial
analysis
malignant
adjacent
non-malignant
tissues
from
five
patients
with
cholangiocarcinoma
or
metastases.
We
find
that
exhibit
recurring,
patient-independent
expression
programs,
reconstruct
ligand–receptor
map
highlights
recurring
tumor–stroma
interactions.
By
combining
transcriptomics
laser-capture
microdissected
regions,
zonation
hepatocytes
sites
characterize
distribution
each
type
across
Our
provides
resource
understanding
malignancies
may
expose
potential
points
interventions.
SYNOPSIS
Single
methods
are
used
generate
microenvironment,
exposing
tumor-stroma
patterns
healthy
tissue.
presented.
Recurring
gene
signatures
found
metastases
cholangiocarcinomas.
Tumor
communicate
through
conserved
ligand-receptor
interaction
network.
Spatial
reveal
zonated
liver.
Introduction
Cancer
heterogeneous
disease,
exhibiting
both
interpatient
intrapatient
variability
(Marusyk
et
al,
2012;
Meacham
Morrison,
2013;
Patel
2014;
Alizadeh
2015).
do
operate
isolation,
rather
closely
interact
complex
milieu
supporting
form
(TME)
(Polyak
2009;
Hanahan
Weinberg,
2011;
Lambrechts
2018).
These
include,
among
others,
range
immune
cells,
cancer-associated
fibroblasts
(CAFs),
endothelial
cells.
Interactions
critical
cancer
survival
(Meacham
2013).
Stromal
supply
factors,
facilitate
evasion,
modulate
composition
extracellular
matrix.
Given
diversity
TME,
it
essential
approaches
resolve
their
(Tirosh
2016;
Puram
2017;
primary
(Llovet
2016).
Tumors
origin
include
hepatocellular
carcinomas
(Guichard
2012),
cholangiocarcinomas
[tumors
originating
cholangiocytes
(Patel,
Sia
2013)],
hepatoblastomas.
Liver
often
originate
colorectal
pancreatic
cause
mortality
(Weinberg,
atlases
provided
important
insight
into
development
(Camp
Segal
2019;
Popescu
2019),
physiology
(MacParland
2018;
Aizarani
pathology
(Zhang
2019,
2020;
Ramachandran
Sharma
2020)
modalities
carcinoma
information,
tissue
identify
distinct
distributions
TME.
Results
To
assemble
analyzed
six
who
underwent
resection
(Fig
1A,
Appendix
Fig
S1).
Three
Patients
hepatic
two
intrahepatic
cholangiocarcinoma,
one
cyst
at
benign
stage
(Dataset
EV1).
dissociated
measured
transcriptomes
using
MARS-seq
(Jaitin
Materials
Methods).
In
parallel,
preserved
microdissection
(LCM)
(Moor
2017,
2018)
molecule
fluorescence
situ
hybridization
(smFISH)
(Bahar
Figure
1.
Experimental
scheme,
tumor,
non-tumor
samples
surgeries
were
scRNA-seq,
frozen
LCM,
fixed
smFISH.
tSNE
plot
colored
normalized
sum
pan-carcinoma
markers
taken
al
(2017).
"n"—indicates
number
per
group.
17
Seurat
clusters
hepatocytes,
(liver
sinusoidal
cells—LSEC,
vascular
cells—LVEC,
cells—LVECt),
mesenchymal
(Stellate
fibroblasts—CAFs,
Pericytes,
smooth
muscle
cells—vSMC),
(Kupffer
scar-associated
macrophages—SAMs,
monocytes
1—TM1,
cDC1,
cDC2,
T
B
cells),
proliferating
Heatmap
showing
marker
genes
(Materials
Expression
maximal
all
types.
Download
figure
PowerPoint
included
7,947
4,140
3,807
1B).
did
show
histological
signs
fibrosis,
exception
patient
p2
Methods,
Dataset
formed
clusters,
which
annotated
based
on
known
recent
cirrhotic
livers
(Ramachandran
2019)
1C).
Notably,
mixture
(Appendix
S1),
demonstrating
signatures.
Cells
several
non-parenchymal
populations—hepatic
stellate
(vSMC),
Kupffer
(LSEC),
(LVEC),
cholangiocytes,
latter
clustering
marked
KRT8,
KRT18,
EPCAM
(Puram
1B)
diverse
TME
populations,
fibroblasts,
Carcinoma
exhibited
differences
metastatic
S1E).
Genes
elevated
higher
cholangiocyte
Beta-defensin
1
(DEFB1)
(Harada
2004)
FGFR2.
metastasis
Cadherin
(CDH17)
(Panarelli
2012)
adhesion
molecules
CEACAM5
CEACAM6,
previously
shown
correlate
colonization
(Powell
extracted
global
unique
1D,
Datasets
EV2
EV3).
validated
panel
12
smFISH
S2).
common
question
whether
reconstructed
stable
regard
numbers
sample
(Mereu
2020).
This
particularly
cancer,
due
profound
levels
heterogeneity
assessed
stability
obtained
our
sampled
end,
mean
subsamples
patients,
equally
sized
as
controls.
compared
subsets
data
those
full
atlas.
gain
correlations,
when
adding
new
strongly
curtailed
most
beyond
three
converged
correlations
subsampling
than
S3).
An
was
cluster,
where
changed
added
thus
demonstrates
that,
while
high
variability,
exhibits
uniform
patients.
Differences
matching
within
same
enabled
identification
populations
compose
2).
von
Willebrand
factor
VWA1,
encoding
glycoprotein
extravasation
(Terraube
2007),
well
SOX17
(Yang
INSR
(Nowak-Sliwinska
promote
angiogenesis
2A).
predominantly
macrophages
(SAMs)
2B).
express
CD9
TREM2,
suppressor
(Tang
CAPG
GPNMB.
GSEA
Subramanian
(2005)
SAM
resulted
significant
enrichment
apical
junction
complement
system.
Their
lipid-associated
genes,
such
PLIN2
LPL,
overlapping
recently
identified
SPP1+
(LAMs)
mouse
fatty
(Remmerie
mononuclear
phagocyte
composed
expressing
C1QB,
MARCO,
CD5L,
CD163
S4).
Tregs,
CTLA4
FOXP3,
whereas
T-cell
cytotoxic
CCL5,
GZMK,
NKG7
2C).
divisions
suggest
recruitment
immune-suppressive
macrophages,
demonstrated
other
(Lambrechts
Binnewies
Additional
conventional
dendritic
(cDC1
cDC2),
(TM1),
FCN1
S100A12
1C,
2.
A–D.
Top-left—tSNE
boxes
demarcate
clusters.
Dashed
labels
indicate
panels
A–C.
(A)
Volcano
differential
(DGE)
samples.
(B)
DGE
phagocytes
(C)
(D)
classified
(cholangiocarcinoma
dark
purple
light
purple).
Wilcoxon
rank-sum
tests
P-values,
Benjamini–Hochberg
multiple
hypotheses
correction
compute
q-values.
Labeled
dots
names
selected
differentially
expressed
further
phagocytes,
2D).
Endothelial
etiologies.
contrast,
up-regulation
chemokines
CCL4,
CCL4L2,
CCL3L3
remodeling
MMP19,
MMP12,
HS3ST2
Diversity
four
3,
S5A).
Hepatic
retinol
binding
protein
(RBP1),
Myosin-11
(MYH11)
abundant
3A,
S1C).
Mesenchymal
Cancer-associated
matrix
(ECM)
COL1A1,
LUM,
BGN,
larger
cluster.
second
cluster
classic
pericytes,
periendothelial
roles
regulating
integrity
(Armulik
2011).
RGS5
CSPG4,
neuron-glial
antigen
2
(NG2)
some
suggested
DES
(Nehls
1992)
ANPEP
(Kumar
2017),
specifically
pericytes
context
Importantly,
almost
absent
demonstrate
RGS5+
indeed
PDGFB,
expected
3B
C).
CAFs
COL1A1
resided
farther
away
D,
3.
A.
Key
(RBP1,
RGS5,
MYH11).
Light
gray
denote
Dark
B.
Left—Representative
image
p1
stained
localization
pericytes.
Scale
bar
10
µm.
lines
mark
shortest
distance
(2a)
(2b)
(1).
Middle—zoom-in
(1)
left
panel,
blood
vessel
like
structure
PDGFB
(magenta)
wrapped
(green).
consecutive
layers
2.5
Right—zoom-in
(2a
b)
distant
signal
RGS5.
DAPI
nuclei
staining.
C,
D.
Violin
vessels
low/high
(n
=
358
n
360,
respectively)
359
359,
respectively).
"p"
P-value
determined
test.
Empty
circles
medians
over
repeats.
E.
Schematic
representation
top-ranked
(bona-fide)
detected
NicheNet
sorted
prior
LVECt
F.
Pathway
bona-fide
EV4)
Enrichr
tool.
Images
representative
images
out
eight
independent
experiments
Paracrine
juxtacrine
attached
be
proper
vascularization
(Annika
2005).
unbiased
signaling
pathways
could
affect
physically
interacting
applied
(Browaeys
computational
method
predicts
induction
downstream
target
3E,
EV4).
via
JAG1,2-NOTCH3,
PDGFB-PDGFRB
S5B),
SLIT2-ROBO1,4
S5C)
ANGPT2-TEK
3E).
ligands
receptors
mediating
cell–pericyte
cross-talk
enriched
pathways,
angiogenesis,
chemokine
cytokine
3F).
highly
dependent
provide
factors
enhance
survival.
turn,
secrete
sensed
(Zhou
2017).
facilitated
an
parsed
database
(Ramilowski
2015)
pairs,
proteins
specific
hand
SAMs
hubs,
representing
49.3%
carcinoma–TME
4A).
focused
recurred
least
4B,
EV5).
resulting
interactome
network
highlighted
modules,
large
module,
modules
centered
around
ERBB,
HGF-MET,
TGFbeta,
FGF,
IGF,
VEGFA,
lipid
trafficking
WNT
planar
polarity
module
4B).
4.
Human
delineates
Summary
total
20%
red
box.
Network
sites.
Node
colors
ligands/receptors
enriched.
Gray
arrows
color
indicates
Zscore
shaded.
Included
significantly
appeared
Dot-plot
highlight
shared
motifs
max
(A).
For
gene,
dot
size
represents
fraction
positive
Top—CAFs
comodulate
carcinoma–stroma
interaction.
produce
DCN
modulates
CAF-SAMs-expressed
ligand
HGF
carcinoma-expressed
receptor
MET.
Bottom—CAFs
CTHRC1
WNT5A
ligand,
CAFs,
SAMS,
TM1
FZD5.
Strategy
computing
scores
tumor.
score
computed
products
average
randomizing
real
manner
preserves
outgoing
incoming
(Scorerand).
ratio
randomized
constitutes
score.
increases
increasing
stage.
Analysis
383
TCGA
(LIHC)
(CHOL).
median
largest
consisted
proteins.
ECM
has
shaped
assembly
degrading
proteins,
collectively
matrisome
(Naba
Varol
Sagi,
Features
stiffness
porosity
optimal
cellular
contacts,
maximize
accessibility
control
exclusion
(Binnewies
Within
produced
collagens
laminins,
integrin
scRNA-seq
identifying
secreting
components
S6).
Planar
(WNT-PCP)
pathway
invasion
(Wang,
2009).
PCP
activated
non-canonical
Wnt
morphogens,
WNT5A,
4B–D).
CTHRC1,
secreted
collagen
triple
helix
filament
forms
stabilizes
its
tumor-expressed
receptor-FZD
(Yamamoto
2008),
CAFs.
Thus,
jointly
WNT-PCP
signaling.
observed
similar
cooperation
MET
module.
driver
(De
Silva
HGF,
activating
MET,
DCN,
decorin
protein,
turn
inhibits
HGF-MET
(Goldoni
revealed
additional
role
interactor
carcinoma-specific
EGFR
summary,
details
connectivity
correlates
severity
convey
selective
advantage
assess
hypothesis,
examined
cohort
bulk-sequenced
4E
F).
first
consists
summed
degree-preserving
random
networks
4E).
normalization
important,
since
simply
reflect
receptors,
oncogenes,
coordinated
receptors.
increased
along
stages
4F).
severity.
identifies
solid
reside
zones
oxygen
levels,
nutrient
availability,
morphogen
concentrations.
can
Itzkovitz,
2017)
result
spatially
organ,
repeating
anatomical
units
termed
lobules,
polarized
centripetal
bloo
Language: Английский
Identification, discrimination and heterogeneity of fibroblasts
Nature Communications,
Journal Year:
2022,
Volume and Issue:
13(1)
Published: June 14, 2022
Abstract
Fibroblasts,
the
principal
cell
type
of
connective
tissue,
secrete
extracellular
matrix
components
during
tissue
development,
homeostasis,
repair
and
disease.
Despite
this
crucial
role,
identification
distinction
fibroblasts
from
other
types
are
challenging
laden
with
caveats.
Rapid
progress
in
single-cell
transcriptomics
now
yields
detailed
molecular
portraits
our
bodies,
which
complement
enrich
classical
histological
immunological
descriptions,
improve
class
definitions
guide
further
studies
on
functional
heterogeneity
subtypes
states,
origins
fates
physiological
pathological
processes.
In
review,
we
summarize
discuss
recent
advances
understanding
fibroblast
how
they
discriminate
types.
Language: Английский
Liver Zonation – Revisiting Old Questions With New Technologies
Rory P. Cunningham,
No information about this author
Natalie Porat‐Shliom
No information about this author
Frontiers in Physiology,
Journal Year:
2021,
Volume and Issue:
12
Published: Sept. 9, 2021
Despite
the
ever-increasing
prevalence
of
non-alcoholic
fatty
liver
disease
(NAFLD),
etiology
and
pathogenesis
remain
poorly
understood.
This
is
due,
in
part,
to
liver’s
complex
physiology
architecture.
The
maintains
glucose
lipid
homeostasis
by
coordinating
numerous
metabolic
processes
with
great
efficiency.
made
possible
spatial
compartmentalization
pathways
a
phenomenon
known
as
zonation.
importance
zonation
normal
function,
it
unresolved
if
how
perturbations
can
drive
hepatic
pathophysiology
NAFLD
development.
While
hepatocyte
heterogeneity
has
been
identified
over
century
ago,
its
examination
had
severely
hindered
due
technological
limitations.
Recent
advances
single
cell
analysis
imaging
technologies
now
permit
further
characterization
cells
across
lobule.
review
summarizes
examining
elucidating
regulatory
role
pathology.
Understanding
organization
metabolism
vital
our
knowledge
provide
targeted
therapeutic
avenues.
Language: Английский
Physiological and pathological roles of lipogenesis
Nature Metabolism,
Journal Year:
2023,
Volume and Issue:
5(5), P. 735 - 759
Published: May 4, 2023
Language: Английский
Understanding tumour endothelial cell heterogeneity and function from single-cell omics
Qun Zeng,
No information about this author
Mira Mousa,
No information about this author
Aisha Shigna Nadukkandy
No information about this author
et al.
Nature reviews. Cancer,
Journal Year:
2023,
Volume and Issue:
23(8), P. 544 - 564
Published: June 22, 2023
Language: Английский