ArticlePDF AvailableLiterature Review

Nieto MA.. The Snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3: 155-166

Authors:

Abstract

The Snail superfamily of zinc-finger transcription factors is involved in processes that imply pronounced cell movements, both during embryonic development and in the acquisition of invasive and migratory properties during tumour progression. Different family members have also been implicated in the signalling cascade that confers left right identity, as well as in the formation of appendages, neural differentiation, cell division and cell survival.
© 2002 Macmillan Magazines Ltd
REVIEWS
Assuming the veracity of Lewis Wolpert’s popular state-
ment
1
that it is not birth, marriage or death, but GASTRU-
LATION
, that is the most important event in the lifespan
of an individual, it seems almost trivial to mention that
the study of
MESODERM formation is a must for develop-
mental biologists. To put it in more conventional terms,
the formation of the third embryonic layer in
TRIPOBLASTIC
animals is, indeed, the time at which the embryonic
axes are coordinated and when the important morpho-
genetic movements that shape the embryo commence.
In this regard, the Snail family of zinc-finger tran-
scription factors occupies a central role in morphogene-
sis, as its members are essential for mesoderm forma-
tion in several organisms from flies to mammals
2–10
.
The analysis of different vertebrate Snail homologues
has highlighted their role not only in the development
of the mesoderm, but also in other processes that
require large-scale cell movements, such as the forma-
tion of the
NEURAL CREST
6,11–14
.
More recently, this role in promoting cell movement
has been extended and includes more generalized phe-
nomena such as the
EPITHELIAL–MESENCHYMAL TRANSITION
(EMT)
15,16
. EMT is the mechanism by which epithelial
cells that are generated in a particular region can disso-
ciate from the epithelium and migrate to reach different
locations
17
. As such, EMT is fundamental to both nor-
mal development and the progression of malignant
epithelial tumours
17
. In addition to triggering EMT,
Snail superfamily members have been implicated in
various important developmental processes, including
neural differentiation, cell fate and survival decisions,
and left–right identity
18
.
From an evolutionary point of view, the Snail fam-
ily provides a good model to study ancestry and the
acquisition of functions that are related to changes in
the
BODY PLAN. In this respect, this family is associated
with the appearance of the neural crest, which is essen-
tial for the formation of the vertebrate head
19
.The
recent identification of new family members and the
association of these members with new functions has
attracted researchers in many fields, from embryonic
pattern formation to cancer research. In this review, I
describe the diversity and organization of the Snail
superfamily, and then address the roles that have been
assigned to the different family members.
The Snail superfamily of repressors
The first member of the Snail family, snail, was
described in Drosophila melanogaster
20,21
, where it was
shown to be essential for the formation of the meso-
derm
2
. Subsequently, Snail homologues have been
found in many species including humans, other verte-
brates, non-vertebrate
CHORDATES (protochordates),
insects,
NEMATODES,ANNELIDS and molluscs (TABLE 1).
Snail family members encode transcription factors
of the zinc-finger type. They all share a similar organi-
zation, being composed of a highly conserved car-
boxy-terminal region, which contains from four to six
zinc fingers, and a much more divergent amino-termi-
nal region. The fingers correspond to the C
2
H
2
type
22
and function as sequence-specific DNA-binding
motifs. The fingers are structurally composed of two
β-strands followed by an α-helix, the amino-terminal
part of which binds to the major groove of the DNA.
THE SNAIL SUPERFAMILY OF ZINC-
FINGER TRANSCRIPTION FACTORS
M. Angela Nieto
The Snail superfamily of zinc-finger transcription factors is involved in processes that imply
pronounced cell movements, both during embryonic development and in the acquisition of
invasive and migratory properties during tumour progression. Different family members have
also been implicated in the signalling cascade that confers left–right identity, as well as in the
formation of appendages, neural differentiation, cell division and cell survival.
GASTRULATION
The morphogenetic movements
of the early embryo that lead to
the generation of the third
embryonic layer — the
mesoderm.
MESODERM
The third embryonic layer
generated during gastrulation,
which occupies an intermediate
position between the ectoderm
and the endoderm. It will give
rise to the skeleton, muscles and
connective tissue.
TRIPOBLAST
An animal that is composed of
three embryonic cell layers:
ectoderm, endoderm and
mesoderm.
NEURAL CREST
A cell population that originates
in the dorsal part of the neural
tube and gives rise to many
derivatives, including most of
the peripheral nervous system,
the cranio-facial skeleton and
pigmented cells of the body.
NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 3 | MARCH 2002 | 155
Instituto Cajal, Doctor Arce,
37, 28002 Madrid, Spain.
e-mail: anieto@cajal.csic.es
DOI: 10.1038/nrm757
© 2002 Macmillan Magazines Ltd
EPITHELIAL–MESENCHYMAL
TRANSITION
The transformation of an
epithelial cell into a
mesenchymal cell with
migratory and invasive
properties.
BODY PLAN
The organization of the
embryonic tissues to generate an
individual with specific
characters.
CHORDATE
An animal with a notochord.
These include ascidians,
amphioxus and all vertebrates.
NEMATODE
An unsegmented worm.
ANNELID
A segmented worm.
BASIC HELIX–LOOP–HELIX
PROTEIN
A transcription factor with a
basic domain that binds to a
hexanucleotide called the E box,
and a hydrophobic domain (the
helix–loop–helix) that allows the
formation of homo- and
heterodimers. They can also
have leucine repeats called a
leucine zipper.
156 | MARCH 2002 | VOLUME 3 www.nature.com/reviews/molcellbio
REVIEWS
mammalian cells
27
. The SNAG domain is conserved in all
vertebrate Snail genes, and is also found in echinoderms,
cephalochordates
7
, in one of the limpet genes
32
and in
Drosophila scratch
33
. Its wide distribution might reflect an
early ancestry. This, in turn, would imply that it has been
lost in other Drosophila family members, Caenorhabditis
elegans and urochordates. Alternatively, the SNAG
domain might have been added independently in each of
the different species. The availability of complete coding
sequences from other groups will help to distinguish
between these two possibilities.
Despite the absence of a SNAG domain, Drosophila
snail also acts as a transcriptional repressor. This activity
is mediated through an interaction with a co-repressor,
CtBP (carboxy-terminal binding protein)
34
. Consensus
motifs for the binding of CtBP are present in other
Drosophila Snail family members (but not scratch) and a
partial consensus is found in several vertebrate family
The two conserved cysteines and histidines (C
2
H
2
)
coordinate the zinc ion. Both random selection and
transfection experiments with different promoters
have shown that the consensus binding site for Snail-
related genes contains a core of six bases,
CAGGTG
15,16,23–26
. This motif is identical to the so-
called E box, the consensus of the core binding site of
BASIC HELIX–LOOP–HELIX
(bHLH) transcription factors,
which indicates that Snail proteins might compete
with them for the same binding sequences
26–28
.
On binding to the E box, Snail family members are
thought to act as transcriptional repressors
9,14–16,24,26,27,29,30
.
The repressor activity depends not only on the finger
region, but also on at least two different motifs that are
found in the amino-terminal region. One of these is the
so-called SNAG (Snail/Gfi) domain, which was initially
described as a repressor domain in the zinc-finger protein
Gfi1
(REF. 31). This motif is important for repression in
Table 1 | Snail superfamily members
Species Common Gene Synonyms Accession no. Map References
name
Caenorhabditis elegans Nematode ces1* AAF01678 I:2.9 37
snail-like K02D7.2 T32983 IV:-26.1 36
scratch-like* C55C2.1 T15225 I:-9.3 36
Helobdella robusta Leech snail1 Hro-sna1 AF410864 43
snail2 Hro-sna2 AF410865 43
Patella vulgata Limpet snail1 Pv-sna1 AY049727 32
snail2 Pv-sna2 AY049791 32
Drosophila melanogaster Fruitfly snail S06222 35D2–3 20,100
escargot AAF12733 35D1 111
worniu S33639 35D2–3 95
scratch* AAA91035 64A2–3 33
scratch-like1* CG12605 AAF47818 64A1 36
scratch-like2* CG17181 AAF47394 61C7 36
Lytechinus variegatus Sea urchin Snail AAB67715 unpublished
Halocynthia roretzi Ascidia Snail BAA75811 8
Ciona intestinalis Ascidia Snail AAB61226 42
Branchiostoma floridae amphioxus Snail AAC35351 7
Takifugu rubripes Pufferfish Snail1 CAB54535 112
Snail2 CAB54536 112
Danio rerio Zebrafish snail1 CAA52795 5,49
snail2 AAA87196 11
slug AI722148 36
scratch* AI883776 36
Xenopus laevis African Snail Xsna P19382 113
clawed toad Slug
α
Xslu AF368041 78,114
Slug
β
Xslu
β
AF368043 78
Silurana tropicalis Western Slug Xslug AF368038 78
clawed frog
Gallus gallus Chicken Snail SnR CAA71033 50,84
Slug CAA54679 6
Mus musculus Mouse Snail Q02085 Chr.2-97.0 3,4
Slug Slugh AAB38365 Chr.16-9.4 50,73,85
Scratch* AY014997 35
Smuc Zfp293 NP038942 26
Homo sapiens Human SNAIL SNAIL1, SNAILH AF155233 20q13.1 115,116
SNAILP SNAI1P AF153502 2q34 115,116
SLUG SLUGH, SNAIL2 AAC34288 8q11 117
SCRATCH1* AY014996 8q24.3 35
SCRATCH2* AL121758 20p12.3–13 35
The Snail superfamily is subdivided into two families: Snail and Scratch (marked by an asterix). Accession numbers are from Entrez
(http://www.ncbi.nlm.nih.gov/Entrez).
© 2002 Macmillan Magazines Ltd
NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 3 | MARCH 2002 | 157
REVIEWS
This proposal is supported by the phylogenetic rela-
tionships that are established when the sequences of the
zinc-finger regions of all Snail superfamily members are
compared
36
. An updated version of such a phylogenetic
tree is shown in
FIG. 1, in which the Scratch genes are
closely grouped and the Snail genes are less tightly asso-
ciated, with several branches that emanate from the base
of the tree. The vertebrate Snail genes seem to be subdi-
vided into two subfamilies that have already been
described: Snail and Slug. The recently isolated mouse
gene Smuc
26
occupies a very unusual position in the
tree, which cannot be easily explained at present. It is
either a gene that originated very early, or it is only pre-
sent in the mouse and has undergone many changes.
Sequence comparisons have allowed the identifica-
tion of consensus sequences for the individual fingers,
both for the Snail and Scratch families as well as a com-
bined consensus for the zinc-finger region of the whole
superfamily
36
(FIG. 2). Signature domains have been
identified in the non-finger region that permit mem-
bers to be ascribed to the Scratch family and the Slug
subfamily
(FIG. 2). On the basis of this phylogenetic
analysis, a model for the evolution of this superfamily
that incorporates the gene duplication events that might
have led to the generation of the family from ancestral
genes is shown in
BOX 1.
Snail in mesoderm and neural-crest formation
In Drososphila embryos, snail is initially expressed in
the prospective mesoderm
38
(FIG. 3),
where it acts as a
repressor to inhibit the expression of neuroectodermal
genes such as rhomboid
39
and single-minded
40
.So,in
Drosophila, mesoderm specification is partly carried
out by the exclusion of alternative cell fates, and snail is
central to this process. The isolation of Snail homo-
logues in different species has confirmed a conserved
role for Snail in mesoderm specification in other
insects
41
, ascidians
8,42
and amphioxus
7
, and mesoderm
development in vertebrates (see below). However, the
expression pattern in the limpet
32
and leech
43
embryos
does not correlate with a role in mesoderm formation,
which indicates that this function cannot be extended
to
LOPHOTROCHOZOANS at present.
In addition to their function in the mesoderm,
vertebrate family members have also been linked
with the development of the neural crest. From an
evolutionary point of view, the appearance of this cell
population is extremely attractive, as, together with
the
EPIDERMAL PLACODES, the neural crest has been crucial
in the formation of the ‘new head’ of vertebrates
19
.
These two tissues differentiate vertebrates from the rest
of the chordates, and their origin correlates with the
shift to active predation and the appearance of paired
sense organs. Indeed, non-vertebrate chordates
(ascidians
8,42
and amphioxus
7
) do not have a neural
crest. However, these chordates do express Snail in
dorsal neural cells, just at the position in which the
neural crest forms in vertebrates
(FIG. 3).So,non-
vertebrate chordates could have the beginnings of a
genetic programme for neural-crest formation, and the
Snail-expressing cells could represent a neural-crest
members. Interestingly, urochordate snail genes, which
lack a SNAG motif, have CtBP consensus sites. So, it is
tempting to speculate that the repressor activity of Snail
proteins has been evolutionarily conserved, but could
use different mechanisms: CtBP co-repression or a
SNAG domain acting alone, or both in conjunction.
A new classification for the Snail family
Recently, new family members have been found in differ-
ent organisms. In particular, several new genes that have
been described in C. elegans, Drosophila, fish, mouse and
human
35,36
are much more similar to Drosophila
scratch
33
and the C. elegans cell death gene ces-1 (REF. 37)
than to any other Snail family member (TABLE 1).This
has led to the proposal that Snail is a superfamily that
can be subdivided into two related but independent
groups: the Snail and the Scratch families
36
.
Figure 1 | Phylogenetic tree of the Snail superfamily. The dark purple square engulfs all the
superfamily members. A light purple background groups the members of the Snail family and a
green background highlights the Scratch family members. The vertebrate Snail and Slug
subfamilies are shown with a light or heavy yellow hatching, respectively. The species shown
represent members of the lophotrochozoans: Pv, Patella vulgata (limpet); ecdysozoans:
Ce, Caenorhabditis elegans (nematode); Dm, Drosophila melanogaster (fruitfly); and
deuterostomes: Bf, Brachiostoma floridae (amphioxus); Ci, Ciona intestinalis (ascidion) and Hr,
Holocynthia roretzi (ascidians); Dr, Danio rerio (zebrafish); Gg, Gallus gallus (chicken), Hs,
Homo sapiens (human); Lv, Lytechinus variegatus (green sea urchin); Mm, Mus musculus
(mouse); Tr, Takifugu rubripes (pufferfish); and Xl, Xenopus laevis (African clawed toad). This is
an updated version of the tree published in REF. 36.
Ce ces1
Mm Smuc
Hr snail
Dm snail
Dm escargot
Dm worniu
Bf snail
Mm
snail
Hs
SNAIL
Hs
SNAILP
XI
Snail
XI Slug
Gg Slug
Mm Slug
Hs Slug
Gg Snail
Pv sna2
Pv sna1
Lv Snail
Dr
snail1
Dr snail2
Tr snail2
Ci snail
Ce snail-like
Dm scratch
Dr scratch
Mm scratch
Hs SCRATCH1
Hs SCRATCH2
Dm scratch-like1
Dm scratch-like2
Ce scatch-like
Snail superfamily
Snail family
Snail subfamily
Vertebrates
Slug subfamily
Scratch family
Tr
snail1
LOPHOTROCHOZOAN
This group includes two
important animal groups, the
Lophophorata (brachiopods, flat
worms and nemerteans) and the
Trochozoa (molluscs and
annelids).
EPIDERMAL PLACODE
An epidermal thickening in the
embryonic head that
differentiates into neurons, as
well as into other cell types, at
the sites at which the sense
organs will form.
© 2002 Macmillan Magazines Ltd
158 | MARCH 2002 | VOLUME 3 www.nature.com/reviews/molcellbio
REVIEWS
in the presence of Slug expression
48
. So, in the chick, Slug
is involved in crest specification all along the anteroposte-
rior axis of the embryo and has an additional role in crest
migration in the head region. It is tempting to speculate
that Snail genes had an ancestral role in the specification
of tissues such as the mesoderm and the neural crest and
that the function in emigration might have been acquired
subsequently. In addition to these data on chick embryos
and the studies in Xenopus that show the role of Slug in
both specification and migration
9,14,45
, the expression pat-
terns of Slug and Snail in other vertebrate embryos, such
as in zebrafish
5,11,49
and mouse
3,4,50
, are also compatible
with their role in neural-crest development.
The role of Snail and Slug in triggering EMT is not
restricted to the mesoderm and neural crest. Snail
and/or Slug are also observed in other cells that undergo
EMT in the developing vertebrate embryo, such as dur-
ing the decondensation of somites
50
, formation of the
parietal endoderm
51
, formation of the heart cushions
52
and closure of the palate (C. Martinez and M.A.N.,
unpublished observations). Even in a mollusc (for exam-
ple the limpet embryo), Snail expression in the involuting
cells of the mantle tips is suggestive of a role in EMT
32
.
This indicates that EMT could be one of the ancestral
functions that are associated with the Snail family.
The function of Snail genes in mesoderm develop-
ment continues after EMT. In ascidians, snail has
been linked with the subdivision of the mesoderm in
precursor population
44
. With respect to vertebrates and
a bona fide neural crest, Slug (a Snail family member)
seems to be involved in neural-crest specification in
both the chick and Xenopus embryos
9,14,45,46
.
Having been specified, both the mesoderm and the
neural crest have to delaminate from the tissue in which
they originate — the
PRIMITIVE STREAK and the neural tube,
respectively — and migrate. Their migration pathways
are well defined, and this enables them to populate diverse
parts of the embryo and contribute to various structures.
Delamination is mediated by the triggering of EMT, and
converts the epithelial cells into mesenchymal cells, which
can migrate through the extracellular matrix
17,47
.
The first indication that the Snail family is involved in
EMT came from studies in the chick embryo. The incu-
bation of early chick embryos with antisense oligonu-
cleotides to Slug inhibited both neural crest and meso-
derm delamination
6
. Defects in crest migration and the
absence of specific crest derivatives have also been
described in Xenopus embryos after Slug antisense treat-
ment
13
or the expression of a dominant-negative Slug
construct
9,14
.Moreover,Slug gain of function leads to an
increase in neural-crest production in the chick embryo
46
.
Interestingly, this increase in the migratory population
was detected only in the head region. Therefore, different
mechanisms operate for neural-crest delamination in the
head and the trunk regions, explaining why inhibition of
neural-crest delamination could occur in the spinal cord
PRIMITIVE STREAK
A structure that is formed at the
posterior end of amniote
embryos at gastrulation stages.
An area of mesoderm
formation.
Figure 2 | Sequence comparison of the main conserved domains and consensus sequences for the individual zinc
fingers of the Snail superfamily. a | Composite of the overall structure of Snail superfamily members, which shows the relative
positions of the SNAG (Snail/Gfi) domain, the zinc fingers (IV), and the Scratch- and Slug-specific boxes. b | Consensus sequences
of the different zinc fingers for the whole superfamily (dark purple) and the Snail (Sna; light purple) and Scratch (Scrt; green) families.
c, d | Sequence comparison of the specific domains that are present in the Slug or Scratch genes, respectively. Dm, Drosophila
melanogaster; Dr, Danio rerio; Gg, Gallus gallus; Hs, Homo sapiens; Mm, Mus musculus; Xl, Xenopus laevis. e | Sequences of the
SNAG domain that are present in representative members of the three big groups of bilateralians. Whereas the zinc-finger region
and the SNAG domain have been shown to be fundamental for protein function, the Slug and Scratch domains represent signature
domains that allow a gene to be unambiguously ascribed to the corresponding family (Scratch) or vertebrate subfamily (Slug).
Abbreviations as above and Bf, Brachiostoma floridae; Lv, Lytechinus variegatus; Pv, Patella vulgata.
--C--C-K-YST--GL-KH---H
--C-ECGK-YATSSNLSRHKQTH
--C--C-K-Y-T---L--H---H
F-CK-C-K-Y-SLGALKMHIRTH
K-CPTC-KAYVSMPALAMH-LTH
--C--C-K-Y-S--AL-MH--TH
C-C--CGKAFSRPWLLQGHIRTH
H-C-VCGK-FSRPWLLQGH-RSH
--C--CGK-FSRPWLLQGH-R-H
F-C-HC-RAFADRSNLRAHLQTH
F-C-HCGKAFADRSNLRAHMQTH
F-C-HC--AFADRSNLRAH-QTH
Y-C--C--TFSRMSLL-KH---G
--C-RC-K-FALKSYL-KH-ES-
--C--C---F---S-L-KH----
Sna I
Scrt I
consensus
Sna II
Scrt II
consensus
Sna III
Scrt III
consensus
Sna IV
Scrt IV
consensus
Sna V
Scrt V
consensus
SDTSS-KDHSGSESPISDEEERLQS-KLSD
SDTSS-KDHSGSESPISDEEERLQP-KLSD
SDTSS-KDHSGSESPISDEEERIQS-KLSD
SDTSS-KDHSGSESPISDEEERLQT-KLSD
SDTSS-KDLSGSESPISDEEERLHT-KLSD
SDTSSNKDHSGSESPRSDEEERIQSTKLSD
Hs SLUG
Mm Slug
Gg Slug
XI Slug α
XI Slug β
Dr slug
AVSEGYAADAFFITDGRSRR
AVTDSYSMDAFFISDGRSRR
AVSEGYAADAFFITDGRSRR
SLSEGYTMDAFFISDGRSRR
AKTVAYTYEAFFVSDGRSKR
Hs SCRATCH1
Hs SCRATCH2
Mm Scratch
Dr Scratch
Dm scratch
MPRSFLVKK
MPRSFLVRK
MPRSFLVKT
MPRSFLIKK
MPRAFLIKK
MPRCLIAKK
All vert. + Lv
Mm, Hs Snail
Mm Smuc
Bf snail
Pv snail 2
Dm scratch
SNAG
domain
Scratch
domain
Slug
domain
Zinc fingers
I II III IV
V
a
b
Zinc-finger consensus sequences
c Slug domain
d Scratch domain
e SNAG domain
© 2002 Macmillan Magazines Ltd
NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 3 | MARCH 2002 | 159
REVIEWS
However, the expression of E-cadherin in the meso-
derm of the Snail mutants is lower than that in the
ectoderm of the same embryos
10
, which indicates that
other cadherin repressors might act simultaneously
with Snail during gastrulation. Candidates include
bHLH-type transcription factors such as SIP1
(REF. 55)
and E47 (REF. 28), which have recently been found to
repress E-cadherin expression, and are also expressed in
the embryonic mesoderm.
A tight regulation of cadherin expression is funda-
mental for the emigration of the neural-crest cells
56,57
.
However, as the Snail-mutant mice die at gastrulation
stages, it has not been possible to address the conse-
quences of Snail loss of function in the neural crest.
Other targets. E-cadherin is the only direct target of
Snail described so far. However, genetic analysis and
overexpression experiments have generated a list of
candidate targets for direct or indirect regulation.
With regard to EMT, in addition to E-cadherin, Snail
transfectants downregulate other epithelial markers,
such as desmoplakin
15
, the epithelial mucin Muc-1
and cytokeratin-18 (
REF. 58; FIG. 4). Mesenchymal
markers such as vimentin and fibronectin are
upregulated and redistributed
15
. These changes can-
not be secondary to the loss of E-cadherin, as trans-
fection of E-cadherin is not enough to induce a
reversion to an epithelial morphology
59
. This indi-
cates that Snail must have additional targets that are
independent of E-cadherin.
different territories
29
— this is in agreement with a
new function described for Slug in Xenopus the pat-
terning of the dorsal mesoderm
9
. Furthermore, the
expression of the fish
5,11,49
, chick
50
and mouse
50
Snail
and Slug, and that of the mouse Smuc gene
26
, also
indicates a role of these proteins in mesodermal pat-
terning and differentiation.
EMT and Snail: target molecules
E-cadherin. The importance of Snail in triggering
EMT in mammals has been confirmed using two
independent approaches. First, Snail was shown to
convert otherwise normal epithelial cells into mes-
enchymal cells through the direct repression of
E-CADHERIN
expression
15,16
. More importantly, Snail knockout ani-
mals die at gastrulation stages and show defects in
EMT
10
. Mutant embryos form a mesodermal layer
that expresses some mesodermal markers, but is com-
posed of columnar cells with apical–basal polarity,
microvilli and
ADHERENS JUNCTIONS, which are all charac-
teristic of epithelial cells
10
. This indicates that they
have failed to undergo EMT. It is known that down-
regulation of E-cadherin is essential for ingression of
the mesodermal cells at gastrulation in mouse
embryos
53
, and in the Snail mutant these cells retain
E-cadherin expression. This is in agreement with Snail
acting as a repressor of E-cadherin expression
15,16
.The
phenotype is reminiscent of that shown by snail
mutants in Drosophila, which also fail to downregu-
late E-cadherin during gastrulation
54
.
METAZOA
The animal kingdom. Includes
sponges, diploblasts,
protostomes and deuterostomes.
PROTOSTOME
An animal in which the mouth
develops from the first opening
that develops in the embryo.
These include ecdysozoans and
lophotrochozoans.
DEUTEROSTOME
An animal in which the anus
develops from the first opening
of the embryo, and the mouth is
formed later. These include
echinoderms and chordates.
E-CADHERIN
The main cell–cell adhesion
molecule, which is central in
maintaining the integrity of
epithelial tissues, both in
physiology and pathology.
ADHERENS JUNCTION
A cell–cell and cell–extracellular
matrix adhesion complex that is
composed of integrins and
cadherins that are attached to
cytoplasmic actin filaments.
Box 1 | Proposed evolutionary history of the Snail gene superfamily
The duplication of a unique snail gene in the
METAZOAN ancestor would have given rise to two genes: snail and scratch.
Independent duplication events in
PROTOSTOMES and DEUTEROSTOMES gave rise to a different number of family members in
each group.
In Drosophila, intra-chromosomal duplications would give rise to three linked genes from each family. Non-vertebrate
chordates seem to have retained the early metazoan situation, with only one gene from each family. This assumes the
existence of a scratch gene that has not yet been isolated.
A whole-genome duplication event proposed to have occurred at the base of the vertebrate lineage
101
, or a massive gene
duplication
102
, would be responsible for the presence of two genes from each family in vertebrates: Snail and Slug on the
one hand, and Scratch1 and Scratch2 on the other hand. Again, an additional, nearly complete genome duplication
103
or
massive local duplications
104
in the teleost (bony fishes) lineage would explain the existence of two very closely related
snail genes (snail1 and snail2) in zebrafish and pufferfish. To distinguish them from ancestral genes, present genes are
shown in bold. Among the latter, the predicted genes are shown in purple.
snail1 snail2 scratch
(ces-1)
scratchscratch-
like
scratch-
like1
scratch-
like2
escargot snail worniu
snail
snail
scratch
snail scratch snail-like scratch snail scratchsnail scratch Snail Scratch
Metazoan ancestor
Protostomes
Lophotrochozoans
Leech and limpet Ascidian and amphioxusC. elegans Drosophila
Ecdysozoans Vertebrates
(Teleosts)
Non-vertebrate chordates
Deuterostomes
snail1 snail2
Snail Slug Scratch1 Scratch2
© 2002 Macmillan Magazines Ltd
160 | MARCH 2002 | VOLUME 3 www.nature.com/reviews/molcellbio
REVIEWS
the neural crest
64
by upregulating Slug
12,64,65
.In Xenopus
and zebrafish, the neural crest is induced at a threshold
concentration of BMP signalling. Higher BMP activity
gives rise to non-neural ectoderm, whereas low (or
null) activity generates neural plate
66,67
. Interestingly,
BMP has been proposed not only as a signal to induce
Slug, but also as a target of it, as overexpression of Slug
induces downregulation of BMPs
9
. Nevertheless, BMP
signalling alone is not sufficient for neural-crest induc-
tion, and studies in Xenopus, zebrafish and mouse have
indicated that members of the Wnt and fibroblast
growth factor (FGF) families are also needed to gener-
ate all the different premigratory precursors
45,68–70
.In
the chick embryo, FGF and BMP cooperate in the gen-
eration of the neural–non-neural boundary — the ter-
ritory of neural-crest specification
71
. So, the combina-
tion of BMP, Wnt and FGF signalling is needed for
neural-crest development.
Given their interactions with BMPs in neural-crest
development, could the FGF and/or Wnt signalling path-
ways induce the expression of Snail family members?
FGF induces Slug expression in extraembryonic epithe-
lial cells
72
and in the rat-bladder-carcinoma cell line
NBT-II
(REF. 73), and upregulates Snail and maintains Slug
expression during limb development in the chick
embryo
74–76
. In addition, mice that have a mutation for
one of the FGF receptors (FGFR1) fail to undergo EMT
at gastrulation, lose Snail expression and show ectopic
expression of E-cadherin
77
in the primitive streak. This
indicates that FGFR1 signalling is needed for the mainte-
nance of Snail expression in the domain of the primitive
streak that is fated to become embryonic mesoderm, and
promotes the downregulation of E-cadherin
77
.
With respect to Wnt signalling, the recent isolation of
Slug promoters in Xenopus has led to the characterization
of a functional binding site for the transcription factor
Lef-1, which regulates gene expression after activation of
Wnt signalling
78
. By contrast, Kwonseop et al.
79
did not
observe Snail or Slug upregulation after overexpression
of LEF in epithelial cells, nor was Snail regulated by LEF
in human colon carcinoma cells
80
. However, an interest-
ing relationship emerges between the FGF and Wnt sig-
nalling pathways through the role of Snail in repressing
E-cadherin expression. Activation of the canonical Wnt
signalling pathway stabilizes β-catenin in the cyto-
plasm, which makes it available to bind the TCF/LEF
transcription factors and together translocate to the
nucleus where they regulate gene expression
81
.
Conversely, high levels of E-cadherin sequester β-
catenin to form adhesion complexes at the cell mem-
brane. So, FGF signalling promotes Wnt signalling by
lowering the levels of E-cadherin through the mainte-
nance of Snail expression. This explains why FGFR1-
mutant mice have attenuated Wnt signalling that can
be reverted by disrupting E-cadherin function
77
.
Another factor that has been shown to induce Snail is
the parathyroid-hormone-related peptide, PTH(rP),
which is essential for triggering the EMT that leads to for-
mation of the
PARIETAL ENDODERM from the PRIMITIVE ENDODERM
and the VISCERAL ENDODERM
51
. This process occurs early in
mouse development, when implantation begins.
Also relevant to EMT is the upregulation of RhoB,
which is important for neural-crest development in
chick embryos
60
, and is ectopically expressed in the chick
neural tube after overexpression of Slug
46
. Regulation of
this small GTPase, which is involved in actin rearrange-
ments, links Snail and EMT with changes in cell shape
and, hence, with the morphogenetic movements that
occur during gastrulation and neural-crest delamina-
tion. Indeed, a Rho-mediated signalling cascade is crucial
for the morphogenetic changes during Drosophila gas-
trulation, a pathway that involves the exchange factor
RhoGEF2 in response to an extracellular signal called
folded gastrulation (Fog)
61
. Considering that, genetically,
Fog lies downstream of Snail
62
, it is tempting to speculate
that Rho GTPases might also be indirect targets of Snail
in the gastrulating fly.
EMT and Snail: inductive signals
Different signalling pathways have been linked with the
induction of Snail family members in the EMT
(FIG. 4).
Transforming growth factor (TGF)-β1 induces EMT
and Snail expression in hepatocytes
63
.TGF-β2 has been
proposed to be a signal for EMT and Slug induction in
heart development
52
; and signalling through other mem-
bers of the TGF-β superfamily — the bone morpho-
genetic proteins (BMPs) — participates in induction of
Figure 3 | Expression of Snail family members in Drosophila, amphioxus, chick and
mouse embryos. In Drosophila, snail is expressed (blue) in the precursors of the mesoderm, and
also later on, when these cells are involuting at gastrulation. In amphioxus, expression is detected
in the mesoderm and at the edges of the neural plate. In vertebrates, the two family members
Snail and Slug are differentially expressed in different species. Note the interchange in the patterns
between chick and mouse. Snail in the mouse and Slug in the chick are expressed in the
precursors of the mesoderm and the neural crest, and also in the migratory populations.
m, mesoderm; nc, neural crest; np, neural plate; pnc, premigratory neural crest; ps, primitive
streak. Photographs of Drosophila and amphioxus embryos have been kindly provided by Maria
Leptin and Jim Langeland, respectively.
snail snail
Snail Slug Snail Slug
Mesoderm Neural crest
Mouse
Chick
Drosophila Amphioxus
m
m
m
m
np
ps
ps
ps
ps
nc
nc
pnc
pnc pnc
pnc
PARIETAL ENDODERM
The extraembryonic tissue that
is derived from the primitive
endoderm and visceral
endoderm, and is composed of
motile cells that secrete high
amounts of extracellular matrix.
PRIMITIVE ENDODERM
The extraembryonic tissue that
gives rise to the visceral and
parietal endoderm.
© 2002 Macmillan Magazines Ltd
NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 3 | MARCH 2002 | 161
REVIEWS
has been explained by the demonstration of an inver-
sion in the expression patterns of Slug and Snail at sites
of EMT
50
. In the chick embryo, Slug is expressed in the
premigratory neural crest and the primitive streak, and
Snail is absent from these tissues; in the mouse embryo,
by contrast, Snail is expressed in these cells, which
undergo EMT
(FIG. 3). This led to the proposal that the
role of Slug in EMT in the chick should be carried out
by Snail in the mouse
50
. The transfection of Snail in
mammalian epithelial cells
15,16
, and the phenotype of
the Snail-mutant mice
10
discussed previously, con-
firmed this prediction. In other vertebrates, the situa-
tion seems more similar to that in the mouse. Indeed,
snail2 is expressed in the premigratory neural crest in
zebrafish
11
, and although both Snail and Slug are
expressed in the premigratory population in Xenopus,
Snail is the first family member to be transcribed
86
.
Experiments that are related to neural-crest develop-
ment in the frog have been carried out only for Slug,so
it will be interesting to analyse the effects of perturbing
Snail function.
The mechanism that is responsible for the observed
interchange is unknown. However, the inversion in
expression sites between chicks and mice is not com-
plete (some sites do not show this change), which indi-
cates that swapping of regulatory modules, differential
loss of tissue-specific cis-regulatory elements or differ-
ential availability of upstream regulators could occur.
Regardless of the mechanism, if Slug induces EMT in
the chick and Snail is responsible in the mouse, are they
functionally equivalent when ectopically expressed at the
appropriate sites? It would be interesting to determine
whether Slug can rescue the gastrulation phenotype
of the mouse Snail mutant. However, there is some
EMT processes also occur during the malignant con-
version of epithelial tumours, and pathological activa-
tion of Snail participates in this process
15,16
(BOX 2)
.The
same signalling molecules seem to operate for the
induction of Snail under these pathological circum-
stances. Indeed, TGF-β induces EMT in epithelial cells
and is necessary for acquisition of the invasive pheno-
type in carcinomas
82,83
. In addition, an integrin-linked
kinase (ILK)-dependent pathway has also been pro-
posed to activate Snail in colon carcinoma cells
80
(FIG. 4).
Different pathways converge in Snail to trigger EMT,
and this places Snail in a central position in this process.
Strict regulation of gene expression is therefore essential
for induction of EMT and maintenance of the migratory
phenotype — an indication of the cooperation that is
required between different signalling cascades. An inter-
esting model, which seems to be in keeping with the
results that have been obtained in different systems, is
that members of the TGF-β/BMP superfamily activate
Snail genes, the levels of which are maintained by FGF
signalling. Snail, in turn, maintains the downregulation
of E-cadherin, and this leaves the Wnt-signalling-medi-
ated, stabilized β-catenin available to bind TCF/LEF pro-
teins and activate gene expression in the nucleus.
Snail and Slug in chick and mouse
Differences in the sites of expression of Snail and Slug
between chick and mouse were the origin of some
confusion. Structural homologues were thought not to
be so, owing to the differences in the expression sites
— indeed, this was the case for the chick Snail-related
(SnR) protein
84
, which is the true Snail homologue
50
.
Studies of Slug-mutant mice showed that Slug is not
essential for mesoderm or neural-crest formation
85
. This
VISCERAL ENDODERM
The extraembryonic cell layer
that is involved in nutrient
uptake and transport.
Figure 4 | Snail genes occupy a central position in triggering EMT in physiological and pathological situations. Different
signalling molecules have been implicated in the activation of Snail genes in several processes that subsequently lead to the
conversion of epithelial cells into mesenchymal cells. Although the action of Snail in the epithelialmesenchymal transition (EMT) as a
direct transcriptional regulator (repressor) has been shown only for E-cadherin, different in vitro and in vivo approaches point to a
series of target genes that are directly or indirectly regulated by these transcription factors. BMP, bone morphogenetic protein; FGF,
fibroblast growth factor; ILK, integrin-linked kinase; PTH(rP)R, parathyroid-hormone-related peptide receptor; TGF-β, transforming
growth factor-β.
FGF
Neural crest
Gastrulation
Epithelial tumour cells
Cytokeratin-18
Epithelial cells
Muc-1
Epithelial
cells
Desmoplakin
Epithelial
cells
E-cadherin
Parietal endoderm
Tumour progression
Hepatocytes
Mesoderm formation
Epithelial cells
Fibronectin
Epithelial cells
Vimentin
Epithelial cells
Rho GTPases
Neural-crest cells
Drosophila gastrulation
Wnt
Neural crest
BMP
Neural
crest
PTH(rP)R
Parietal
endoderm
Integrin
Tumour cells
TGF-β
Tumour progression
Hepatocytes
Heart development
Palatal fusion
Snail/Slug
ILK
Epithelial markers Mesenchymal markers Cytoskeletal changes
© 2002 Macmillan Magazines Ltd
162 | MARCH 2002 | VOLUME 3 www.nature.com/reviews/molcellbio
REVIEWS
leukaemic factor; a putative CES-2 homologue) for the
E2A-positive transactivation domain. This leads to acti-
vation of a different family member, Slug, which in turn
represses a partial homologue of EGL-1 (BH3) and ren-
ders the anti-apoptotic BCL-X
L
protein active to pro-
mote survival, leading to leukaemia
25
(FIG. 5). So, both
Scratch and Slug seem to function as anti-apoptotic
agents, in agreement with data on the regulation of Slug
during limb development in the chick
75,76
.Here,Slug is
downregulated in the areas that are destined to die, and
is proposed to act as a survival factor that maintains the
undifferentiated mesenchymal phenotype.
Cell division and endoreduplication
During Drosophila gastrulation, the changes in cell
shape that are associated with formation of the ventral
furrow are accompanied by inhibition of mitosis. This
links morphogenesis with cell division. This inhibition
is mediated by Tribbles, a serine/threonine kinase that
counteracts String, the homologue of the
CDC25 phos-
phatase that is necessary for mitosis
87,88
. This inhibition
functional equivalence, at least during embryonic devel-
opment, both within and between species. Ectopic
expression of chick and mouse Snail in the chick hind-
brain induces an increase in neural-crest production, in
a similar way to that of the endogenous gene, Slug
46
.But
it is not clear whether this functional equivalence also
occurs during tumour progression, as Slug is expressed
in different carcinoma-derived cell lines regardless of
their phenotype in terms of
INVASIVENESS
15
.
Snail superfamily and cell survival
Several lines of evidence point to a role for Snail
superfamily members in regulating cell death or sur-
vival. In a particular population of C. elegans neurons,
the protein involved in cell death, CES-2, represses
CES-1 (scratch) function. This allows the cell-death
activator EGL-1 to repress the survival gene ced-9, and
allows the action of the cell-death proteins CED-4 and
CED-3 (
REF. 37; FIG. 5).
In some human leukaemias, a chromosomal translo-
cation swapped the repression domain of HLF (hepatic
INVASIVENESS
The ability to degrade and
migrate through the
extracellular matrix.
Box 2 | The epithelial–mesenchymal transition in tumour progression
The epithelial–mesenchymal
transition (EMT) occurs not
only during normal
embryonic development, but
also in pathological situations
such as acquisition of the
invasive phenotype in
epithelial tumours, in which it
constitutes the first step for
the formation of metastasis.
This pathological EMT has
been associated with the
downregulation of E-cadherin
expression and the acquisition
of migratory properties.
Indeed, the loss of E-cadherin
expression is crucial for the
progression from adenoma to
carcinoma
105
.
The idea that pathological
activation of Snail genes
could be involved in tumour
progression was proposed
several years ago
6
, and has
been shown recently. Snail is
a strong direct repressor of
E-cadherin expression, and
Snail transfection confers
tumorigenic, invasive and
migratory properties to
otherwise normal epithelial
cells
15,16
. An inverse
correlation has been found
between Snail and E-cadherin expression in mouse and human cell lines
15,16,106,107
. Furthermore, Snail is activated in vivo
at the invasive front of chemically induced mouse skin tumours
15
, and it is present in human breast carcinomas
108,109
,in
which it inversely correlates with the degree of differentiation and is associated with lymph-node metastasis
109
.As such,
Snail can be now considered a marker of malignancy; this paves the way for the design of anti-invasive therapies and
makes the search for endogenous or artificial regulators of exceptional interest.
Mesoderm formation
Invasive area in primary tumour
Neural-crest delamination
Early mesoderm
Primitive streak
Neural tube
Neural
crest
Epithelial cells
Mesenchymal cells
(invasive)
Snail
Invasive cells Tumour cells
Break point of
basement
membrane
Basement membrane
Snail-negative cells Snail-positive cells
Embryonic development
Snail transfection Tumour progression
Epithelial–mesenchymal transition
© 2002 Macmillan Magazines Ltd
NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 3 | MARCH 2002 | 163
REVIEWS
Snail in left–right asymmetry
A striking asymmetric and transient Snail expression
in the right-hand lateral mesoderm of the chick
embryo led Cooke and colleagues
84
to investigate a
possible role for this gene in the establishment of
LEFT–RIGHT ASYMMETRY. Incubation of early chick
embryos with antisense oligonucleotides to Snail led
to a randomization of the
HEART SITUS
84
. Further exper-
iments
93,94
established that Snail lies in the genetic
cascade that gives rise to bilateral body asymmetries.
Snail is downstream of the signal that is generated by
the TGF-β superfamily member Nodal, and
upstream of the transcription factor Pitx-2 — a
bicoid-type homeobox protein that is responsible for
activating the left-side-specific differentiation pro-
gramme
(FIG. 5). Inhibition of the BMP signal that
inactivates Nodal on the left side of the embryo leads
to the repression of Snail, which in turn cannot
repress Pitx-2. The left–right asymmetric expression
of Snail is also observed in the mouse embryo, which
constitutes one of the few sites of mesodermal
expression that have not been interchanged between
chick and mouse at these early stages
50
. The transient
nature of this asymmetric expression, particularly in
the mouse, might explain why it has not been
detected in Drosophila or other vertebrates. It would
be interesting to re-analyse other species, as a
left–right asymmetric expression has also been found
in the limpet Patella vulgata for one of the two Snail
genes isolated, sna2
(REF. 32). This conservation indi-
cates that this might be an ancient function that is
associated with the Snail family.
depends on Snail function
88
, which, therefore, might
act as a mitotic inhibitor. This is in agreement with
the low proliferation rate that is observed in Snail-
transfected epithelial cells compared with control cells
(S. Vega and M.A.N., unpublished observations). It
seems reasonable that cells that undergo massive
cytoskeletal reorganization associated with changes in
cell shape or active migration are prevented from
undergoing cell division.
Other members of the Snail family — including
mouse Snail itself — have been associated with mitosis
in two processes. Indeed, Escargot and mouse Snail are
involved in the control of polyploidy in several tissues,
including
IMAGINAL DISCS cells in Drosophila
24,89
, mouse
TROPHOBLAST cells
27
and human megakaryocytes
90
. Both
proteins inhibit
ENDOREDUPLICATION, and therefore induce
progression of the cell cycle to mitosis. The molecular
mechanism could be related to the activation of String,
as this protein is involved in the control of mitosis cou-
pled to the process of
ASYMMETRIC CELL DIVISION in
Drosophila
91
(FIG. 5). Certainly, a deficiency in the three
Snail-family members (snail, escargot and worniu) leads
to an inappropriate activation of Inscutable, which con-
trols the subcellular localization of Prospero, a key pro-
tein in determining the
GANGLION MOTHER CELL fate
91,92
.So,
depending on the cellular process, at least in Drosophila,
Snail seems to act as an inhibitor or an activator of
String. Snail transcription factors have been shown to
act as repressors
9,14–16,24,26,27,29,30
, which indicates that
Snail-mediated activation could be the result of an indi-
rect regulation. However, the possibility that they act as
activators cannot be excluded at the moment
18
.
CDC25
A family of protein phosphatases
that dephosphorylate cyclin-
dependent kinases during cell-
cycle progression.
IMAGINAL DISCS
The primordia of different adult
structures that are present in the
larvae of insects with complete
metamorphosis.
TROPHOBLAST
The extraembryonic epithelial
tissue that is crucial for
formation of the placenta.
ENDOREDUPLICATION
The process by which the cells
pass to rounds of DNA
duplication in the absence of a
mitotic division.
ASYMMETRIC CELL DIVISION
A process by which a cell gives
rise to two different descendants
after division.
GANGLION MOTHER CELL
One of the daughters of a
Drosophila neuroblast after
asymmetric cell division. It
divides once more to give rise to
two post-mitotic neurons.
LEFT–RIGHT ASYMMETRY
The differences along the
left–right axis of the body.
HEART SITUS
The position of the heart with
respect to the left–right axis of
the body.
Figure 5 | Different genetic pathways involving Snail function. In addition to triggering the epithelialmesenchymal transition,
Snail function has been described in several genetic pathways that lead to a | cell death or survival, b | asymmetric cell division and
c | leftright (L/R) asymmetry. In all cases, arrows indicate the flow of the pathway, not direct transcriptional repression or activation.
Although function as transcriptional activators cannot be fully excluded, Snail proteins have been described as transcriptional
repressors in all the species analysed so far. To follow the sequence of active proteins in the corresponding pathway, genes that are
repressed or inactive are shown in red, and the inactive regulatory steps are shown as dotted lines. HLF, hepatic leukaemic factor;
BMP, bone morphogenetic protein; FGF, fibroblast growth factor.
C. elegans/human Human leukaemia Vertebrates
CES-2/HLF?
EGL-1/BH3
CED-9/BCL-X
L
CED-4/APAF-1
CED-3/caspase-9
Cell death
Left Right
CES-1/Slug
(Scratch)
Drosophila
Proneural genes
Inscutable String (cdc25)
Neuroblast
asymmetry
Neuroblast
division
Prospero asymmetric
localization
Ganglion mother-cell fate
Snail genes
(snail, escargot, worniu)
E2A-HLE (HLF con-
verted in activator)
BH3
BCL-X
L
Cell survival
(leukaemogenesis)
Slug
BMP
antagonist
Nodal
Snail
Pitx2
BMP
Nodal
Snail
Pitx2
BMP
FGF8
Cell survival L/R asymmetry
Neuroblast asymmetric
cell division
abc
© 2002 Macmillan Magazines Ltd
164 | MARCH 2002 | VOLUME 3 www.nature.com/reviews/molcellbio
REVIEWS
However, this function is not unique for this family,
as vertebrate Snail and Slug are expressed in the ner-
vous system at later developmental stages (F. Marin
and M.A.N., unpublished observations), indicating
that, probably, neuronal differentiation might be a
function that is associated with both the Snail and
Scratch families
(BOX 3).
Cooperativity and antagonism
What is the relationship between different members of
the Snail superfamily when they act in the same biologi-
cal process or on similar targets? Interestingly, there are
examples of both cooperativity and antagonism.
With respect to cell differentiation, chick Slug
and mouse Snail and Slug have been proposed to
maintain the mesenchymal phenotype and repress dif-
ferentiation
15,75,76
. Similarly, Drosophila snail and escargot
also maintain the undifferentiated phenotype — they
antagonize neurogenesis by competing with bHLH pro-
teins
97
. So, they seem to antagonize the role of scratch in
promoting neural differentiation in Drosophila
33
, C. ele-
gans
37
and mouse
96
.
Chick and human Slug are associated with cell sur-
vival
25,75,76
, whereas chick Snail has been associated with
the apoptotic programme in the developing limb
74
.
With regard to target genes, Snail represses E-cadherin
expression during mesoderm formation in Drosophila
54
and mammals
15,16
; by contrast, escargot activates cad-
herin expression during tracheal development in the fly.
In some cases, different family members cooperate,
such as the three Drosophila snail genes in neurogene-
sis
95
and asymmetric cell division
91,92
.Moreover,snail
and escargot cooperate in wing development
99
in the fly,
and the vertebrate Snail and Slug genes might also coop-
erate in triggering EMT and maintaining the mesenchy-
mal phenotype during neural-crest development
13–15
.
Finally, a striking example is the regulation of
String by Drosophila snail, which seems to activate it
during asymmetric cell division
91
and inhibit it dur-
ing gastrulation
88
.
Perspectives
Although we now have invaluable information on the
different processes in which the Snail superfamily pro-
teins are involved — both during development and in
some pathological situations — we are a long way from
fully understanding their functions and mutual relation-
ships. Further work will take advantage of the completed
genomes and of the new imaging approaches that allow
cell movements to be followed in the living embryo.
As Snail-mutant mice die at gastrulation, spatio-
temporal, conditional Snail-mutant mice are needed to
study the participation of Snail in later processes such as
formation of the neural crest or differentiation of tissues
and organs, including the mesoderm. In terms of the
role of Snail in the appearance of the neural crest during
evolution, experiments that are similar to those carried
out for the Hox genes — in which regulatory sequences
from non-vertebrate chordates are introduced in trans-
genic mice
100
— will help to challenge the genetic pro-
gramme that is already present in the proposed precursor
The Snail superfamily in neural development
Although the four Drosophila genes that have been
analysed so far — snail, escargot, worniu and scratch
are prominently expressed in the nervous system, indi-
vidual mutants do not show a strong neural phenotype.
However, double mutants of scratch and the HLH pro-
tein deadpan show loss of neurons
33
. Similarly, deletion
of snail, escargot and worniu leads to the loss of central
nervous system determinants
95
. The identification of
two additional scratch-related genes in Drosophila
36
indi-
cates that the three scratch genes could collaborate, as
the Snail members do, and a strong neural phenotype
might be expected for the triple mutant.
Interestingly, the C. elegans scratch homologue ces-1
(REF. 37) is essential for the formation of neurons, and
the mouse Scratch gene is neural specific and induces
neuronal differentiation in P19 embryonal carcinoma
cells
96
.In addition,a Scratch homologue is specifically
expressed in the primary neurons of the zebrafish
embryo (M. J. Blanco and M.A.N., unpublished obser-
vations), which indicates a neuronal-specific function
for both the invertebrate and vertebrate Scratch family.
Box 3 | Proposed ancestral and derived functions of the Snail superfamily
Phylogenetic and expression studies together with functional analyses in different model
organisms allow ancestral and acquired functions to be proposed for the different Snail
superfamily groups in metazoan evolution. An ancestral function in the development of
sensory and/or neuronal structures is proposed for the whole superfamily
36
,which
includes both the Snail and Scratch genes.An additional ancestral function in the control
of cell death/survival is also proposed
25,37,75,76
.
The role in epithelial–mesenchymal transitions (EMTs) seems to be exclusively
associated with the members of the Snail family, with representatives analysed in
Lophotrochozoans,
ECDYSOZOANS and deuterostomes. This role in EMT has been co-opted
for cell migration during mesoderm and neural-crest formation and tumour
progression, when these processes emerged
36
. In vertebrates, Snail and/or Slug proteins
participate in this process depending on the species. Further roles in the development of
appendages
74–76,99
and cell division
24,27,88–92
are associated with particular members of the
Snail family in different groups. Finally, a still-uncharacterized role in lens development
50
has been specifically proposed for Slug subfamily members, which seem to participate
neither in cell division nor in the definition of left–right (L/R) asymmetry. None of the
known functions has been specifically associated with the Scratch family or the
vertebrate Snail subfamily.
Wing/limb
development
L/R asymmetry
Mesoderm
specification/
development
Neural-crest
specification
Neural-crest
delamination
Lens developmentSlug
Snail family
(includes Slug
in vertebrates)
Cell division and EMT
Neural/sensory
development
Cell survival
Snail superfamily
(snail + scratch)
Metazoan
ancestor
Arthropods
and/or molluscs
Non-vertebrate
chordates
Vertebrates Mammals
Tumour
progression
ECDYSOZOANS
One of the important groups
within the animal kingdom, it
includes arthropods and
nematodes.
© 2002 Macmillan Magazines Ltd
NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 3 | MARCH 2002 | 165
REVIEWS
transcription factors for binding to E boxes will depend
on relative affinities that might need the participation
of different co-regulators, and cooperation with bHLH
proteins or other unidentified partners could provide
additional degrees of complexity for the patterning and
differentiation of specific cell types.
The description of Scratch as a new family offers
unexplored territory for the study of new functions in
the different species. And finally, the implication of the
Snail family in pathology challenges the use of
amenable systems to identify specific repressors that can
be used to develop new therapeutic strategies.
population
44
. Obviously, characterization of the regula-
tory sequences that drive specific spatio-temporal
expression of the different members in different tissues
and species is a long-term goal that has to be
approached systematically.
From a more biochemical point of view, we have lit-
tle information on the mechanism that is used by Snail
for transcriptional regulation. We do not know whether
Snail genes can act as activators, and have little infor-
mation on the proteins that induce or repress their
expression, the targets they regulate or the nature of
the transcription complex. Competition with bHLH
1. Wolpert, L. Quoted in Slack, J. M. W. From Egg to Embryo:
Determinative Events in Early Development, Vol. 1
(Cambridge Univ. Press, Cambridge, UK, 1986).
2. Alberga, A., Boulay, J. L., Kempe, E., Dennefield, C. &
Haenlin, M. The snail gene required for mesoderm formation
in Drosophila is expressed dynamically in derivatives of all
three germ layers. Development 111, 983992 (1991).
3. Nieto, M. A., Bennet, M. F., Sargent, M. G. & Wilkinson, D. G.
Cloning and developmental expression of Sna, a murine
homologue of the Drosophila snail gene. Development 116,
227237 (1992).
4. Smith, D. E., Del Amo, F. F. & Gridley, T. Isolation of Sna, a
mouse gene homologous to the Drosophila genes snail and
escargot: its expression pattern suggests multiple roles
during postimplantation development. Development 116,
10331039 (1992).
5. Hammerschmidt, M. & Nüsslein-Volhard, C. The expression
of a zebrafish gene homologous to Drosophila snail suggests
a conserved function in invertebrate and vertebrate
gastrulation. Development 119, 11071118 (1993).
6. Nieto, M. A., Sargent, M., Wilkinson, D. G. & Cooke, J.
Control of cell behaviour during vertebrate development by
Slug, a zinc finger gene. Science 264, 835839 (1994).
The isolation of the first Slug gene and its involvement
in delamination of the neural crest and of the early
mesoderm.
7. Langeland, J. A., Tomsa, J. M., Jackman, W. R. Jr & Kimmel,
C. B. An amphioxus snail gene: expression in paraxial
mesoderm and neural plate suggests a conserved role in
patterning the chordate embryo. Dev. Genes Evol. 208,
569577 (1998).
8. Wada, S. & Saiga, H. Cloning and embryonic expression of
Hrsna, a snail family gene of the ascidian Halocynthia roretzi:
implication in the origins of mechanisms for mesoderm
specification. Dev. Growth Differ. 41, 918 (1999).
9. Mayor, R., Guerrero, R., Young, R. M., Gomez-Skarmeta,
J. L. & Cuellar, C. A novel function for the Xslug gene: control
of dorsal mesendoderm development by repressing BMP-4.
Mech. Dev. 97, 4756 (2000).
10. Carver, E. A., Jiang, R., Lan, Y., Oram, K. F. & Gridley, T.
The mouse Snail gene encodes a key regulator of the
epithelialmesenchymal transition. Mol. Cell. Biol. 21,
81848188 (2001).
This study shows that Snail-mutant mice die at
gastrulation owing to defective EMT.
11. Thisse, C., Thisse, B. & Postlethwait, J. H. Expression of
snail2, a second member of the zebrafish snail family, in
cephalic mesendoderm and presumptive neural crest of
wild-type and spadetail mutant embryos. Dev. Biol. 172,
8699 (1995).
12. Mancilla, A. & Mayor, R. Neural crest formation in Xenopus
laevis: mechanisms of Slug induction. Dev. Biol. 177,
580589 (1996).
13. Carl, T. F., Dufton, C., Hanken, J. & Klymkowsky, M. W.
Inhibition of neural crest migration in Xenopus using
antisense slug RNA. Dev. Biol. 213, 101115 (1999).
14. LaBonne, C. & Bronner-Fraser, M. Snail-related
transcriptional repressors are required in Xenopus for both
the induction of the neural crest and its subsequent
migration. Dev. Biol. 221, 195205 (2000).
15. Cano, A. et al. The transcription factor snail controls
epithelialmesenchymal transitions by repressing E-cadherin
expression. Nature Cell Biol. 2, 7683 (2000).
This study shows that Snail triggers EMT through the
repression of E-cadherin transcription. This also
shows the correlation of Snail expression with the
invasive phenotype in cell lines and in tumours
induced in the skin of the mouse.
16. Batlle, E. et al. The transcription factor snail is a repressor of
E-cadherin gene expression in epithelial tumour cells. Nature
Cell Biol. 2, 8489 (2000).
The demonstration that Snail is a direct repressor of
E-cadherin expression in tumour cells.
17. Hay, E. D. An overview of epithelio-mesenchymal
transformation. Acta Anat. 154, 820 (1995).
18. Hemavathy, K., Ashraf, S. I. & Ip, Y. T. Snail/Slug family of
repressors: slowly going to the fast lane of development and
cancer. Gene 257, 112 (2000).
19. Gans, C. & Northcutt, R. G. Neural crest and the evolution of
vertebrates: a new head. Science 220, 268274 (1983).
20. Grau, Y., Carteret, C. & Simpson, P. Mutations and
chromosomal rearrangements affecting the expression of
snail, a gene involved in embryonic patterning in Drosophila
melanogaster. Genetics 108, 347360 (1984).
21. Nusslein-Volhard, C., Weischaus, E. & Kluding, H. Mutations
affecting the pattern of the larval cuticle in Drosophila
melanogaster. I. Zygotic loci on the second chromosome.
Wilheim Roux’s Arch. Dev. Biol. 193, 267282 (1984).
References 20 and 21 simultaneously described the
first Snail family member.
22. Knight, R. & Shimeld, S. Identification of conserved C
2
H
2
zinc-finger gene families in the Bilateralia. Genome Biol. 2,
research0016.10016.8 (2001).
23. Mauhin, V., Lutz, Y., Dennefeld, C. & Alberga, A. Definition of
the DNA-binding site repertoire for the Drosophila
transcription factor SNAIL. Nucleic Acids Res. 21,
39513957 (1993).
24. Fuse, N., Hirose, S. & Hayashi, S. Diploidy of Drosophila
imaginal cells is maintained by a transcriptional repressor
encoded by escargot. Genes Dev. 8, 22702281 (1994).
The first indication of a Snail family member that is
involved in endoreduplication.
25. Inukai, T. et al. Slug, a ces-1-related zinc finger transcription
factor gene with antiapoptotic activity is a downstream target
of the E2A-HLF oncoprotein. Mol. Cell 4, 343352 (1999).
This study provides a description of the anti-apoptotic
effect of Slug in humans.
26. Kataoka, H. et al. A novel snail-related transcription factor
Smuc regulates basic helixloophelix transcription factor
activities via specific E-box motifs. Nucleic Acids Res. 28,
626633 (2000).
27. Nakayama, H., Scott, I. C. & Cross, J. C. The transition to
endoreduplication in trophoblast giant cells is regulated by
the mSna zinc finger transcription factor. Dev. Biol. 199,
150163 (1998).
28. Pérez-Moreno, M. et al. A new role for E12/E47 in the
repression of E-cadherin expression and
epithelialmesenchymal transition. J. Biol. Chem. 276,
2742427431 (2001).
29. Fijiwara, S., Corbo, J. C. & Levine, M. The snail repressor
establishes a muscle/notochord boundary in the Ciona
embryo. Development 125, 25112520 (1998).
30. Hemavathy, K., Guru, S. C., Harris, J., Chen, J. D. & Ip, Y. T.
Human Slug is a repressor that localizes to sites of active
transcription. Mol. Cell. Biol. 26, 50875095 (2000).
31. Grimes, H. L., Chan, T. O., Zweidler-McKay, P. A., Tong, B. &
Tsichlis, P. N. The Gfi1 proto-oncoprotein contains a novel
transcriptional repressor domain, SNAG, and inhibits G
1
arrest induced by inteleukin-2 withdrawal. Mol. Cell. Biol. 16,
62636272 (1996).
32. Lespinet, O. et al. Characterization of two snail genes in the
gastropod mollusk Patella vulgata. Implications for
understanding the ancestral function of the snailrelated
genes in Bilateralia. Submitted.
This study links Snail genes with EMT and leftright
asymmetry in molluscs.
33. Roark, M., Sturtevant, M. A., Emery, J., Vaessin, H. & Grell, E.
scratch, a pan-neural gene encoding a zinc finger protein
related to snail, promotes neural development. Genes Dev. 9,
23842398 (1995).
34. Nibu, Y. et al. dCtBP mediates transcriptional repression by
Knirps, Kruppel and Snail in the Drosophila embryo. EMBO
J. 17, 70097020 (1998).
35. Nakakura, E. K. et al. Mammalian Scratch: a neural-specific
Snail family transcriptional repressor. Proc. Natl Acad. Sci.
USA 98, 40104015 (2001).
36. Manzanares, M., Locascio, A. & Nieto, M. A. The increasing
complexity of the Snail superfamily in metazoan evolution.
Trends Genet. 17, 178181 (2001).
The organization and new classification for members
of the Snail superfamily.
37. Metzstein, M. M. & Horwitz, H. R. The C. elegans cell death
specification gene ces-1 encodes a Snail family zinc finger
protein. Mol. Cell 4, 309319 (1999).
The identification of the first Snail family member in
nematodes.
38. Leptin, M. twist and snail as positive and negative regulators
during Drosophila mesoderm development. Genes Dev. 5,
15681576 (1991).
39. Ip, Y. T., Park, R., Kosman, D. & Levine, M. The dorsal
gradient morphogen regulates stripes of rhomboid
expression in the presumptive neruoectoderm of the
Drosophila embryo. Genes Dev. 6, 17281739 (1992).
40. Kasai, Y., Nambu, J. R., Lieberman, P. M. & Crews, S. T.
Dorsalventral patterning in Drosophila: DNA binding of snail
protein to the single-minded gene. Proc. Natl Acad. Sci. USA
89, 34143418 (1992).
41. Somer, R. J. & Tautz, D. Expression patterns of twist and
snail in Tribolium (Coleoptera) suggest a homologous
formation of mesoderm in long and short germ band insects.
Dev. Genet. 15, 3237 (1994).
42. Corbo, J. C., Erives, A., Di Gregorio, A., Chang, A. & Levine, M.
Dorsoventral patterning of the vertebrate neural tube is
conserved in a protochordate. Development 124,
23352344 (1997).
The isolation of the first snail gene in a non-vertebrate
chordate.
43. Goldstein, B., Leviten, M. W. & Weisblat, D. A. Dorsal and
Snail homologs in leech development. Dev. Genes Evol. 211,
329337 (2001).
The isolation of the first Snail homologue in
lophotrochozoans.
44. Holland, L. Z. & Holland, N. D. Evolution of neural crest and
placodes: amphioxus as a model for the ancestral
vertebrate? J. Anat. 199, 8598 (2001).
Although Slug-mutant mice do not show gross
abnormalities in either neural crest or mesoderm
formation, further analysis of these mutants will
provide information on the specific roles of Slug in
mammals in these and other tissues.
45. LaBonne, C. & Bronner-Fraser, M. Neural crest induction in
Xenopus: evidence for a two-signal model. Development
125, 24032414 (1998).
46. Del Barrio, M. G. & Nieto, M. A. Overexpression of Snail family
members highlights their ability to promote chick neural crest
formation. Development 129, 15831594 (2002).
47. Erickson, C. A. in Principles of Tissue Engineering, 2nd Edn
1931 (Academic, San Diego, USA, 2000).
48. Sela-Donenfeld, D. & Kalcheim, C. Regulation of the onset of
neural crest migration by coordinated activity of BMP4 and
Noggin in the dorsal neural tube. Development 126,
47494762 (1999).
49. Thisse, C., Thisse, B., Schilling, T. F. & Postlethwait, J. H.
Structure of the zebrafish snail1 gene and its expression in
wild-type, spadetail and no tail mutant embryos.
Development 119, 12031215 (1993).
50. Sefton, M., Sanchez, S. & Nieto, M. A. Conserved and
divergent roles for members of the Snail family of
transcription factors in the chick and mouse embryo.
Development 125, 31113121 (1998).
51. Velmaat, J. M. et al. Snail an immediate early target gene of
parathyroid hormone related peptide signaling in parietal
endoderm formation. Int. J. Dev. Biol. 44, 297307 (2000).
© 2002 Macmillan Magazines Ltd
166 | MARCH 2002 | VOLUME 3 www.nature.com/reviews/molcellbio
REVIEWS
52. Romano, L. & Runyan, R. B. Slug is an essential target of
TGFβ2 signaling in the developing chicken heart. Dev. Biol.
223, 91102 (2000).
53. Burdsal, C. A., Damsky, C. H. & Pedersen, R. A. The role of
E-cadherin and integrins in mesoderm differentiation and
migration at the mammalian primitive streak. Development
118, 829844 (1993).
54. Oda, H., Tsukita, S. & Takeichi, M. Dynamic behavior of the
cadherin-based cellcell adhesion system during Drosophila
gastrulation. Dev. Biol. 203, 435450 (1998).
55. Comijn, J. et al. The two-handed E box binding zinc finger
protein SIP1 downregulates E-cadherin and induces
invasion. Mol. Cell 7, 12671278 (2001).
56. Nakagawa, S. & Takeichi, M. Neural crest cellcell adhesion
controlled by sequential and subpopulation-specific
expression of novel cadherins. Development 121,
13211332 (1995).
57. Nakagawa, S. & Takeichi, M. Neural crest emigration from
the neural tube depends on regulated cadherin expression.
Development 125, 29632971 (1998).
58. Garcia de Herreros, A. in Common Molecules in
Development and Carcinogenesis (Juan March Foundation,
Madrid, Spain, 2001).
59. Navarro, P., Lozano, E. & Cano, A. Expression of E- or P-
cadherin is not sufficient to modify the morphology and the
tumorigenic behavior of murine spindle carcinoma cells.
Possible involvement of plakoglobin. J. Cell Sci. 105,
923934 (1993).
60. Liu, J. P. & Jessell, T. M. A role for rhoB in the delamination of
neural crest from the dorsal neural tube. Development 125,
50555067 (1998).
61. Barrett, K., Leptin, M. & Settleman, J. The Rho GTPase and
a putative RhoGEF mediate a signaling pathway for the cell
shape changes in Drosophila gastrulation. Cell 91, 905915
(1997).
62. Morize, P., Christiansen, A. E., Costa, M., Parks, S. &
Wieschaus, E. Hyperactivation of the folded gastrulation
pathway induces specific cell shape changes. Development
125, 589597 (1998).
63. Spagnoli, F. M., Cicchini, C., Tripodi, M. & Weiss, M. C.
Inhibition of MMH (Met murine hepatocyte) cell differentiation
by TGF-β is abrogated by pre-treatment with the heritable
differentiation effector FGF1. J. Cell Sci. 113, 36393647
(2000).
64. Liem, K. F. Jr, Tremml, G., Roelink, H. & Jessell, T. Dorsal
differentiation of neural plate cells induced by BMP-mediated
signals from epidermal ectoderm. Cell 82, 969979 (1995).
65. Dickinson, M. E., Selleck, M. A., McMahon, A. P. & Bronner-
Fraser, M. Dorsalization of the neural tube by the non-neural
ectoderm. Development 121, 20992106 (1995).
66. Marchant, L., Linker, C., Ruiz, P. & Mayor, R. The inductive
properties of mesoderm suggest that the neural crest cells
are specified by a gradient of BMP. Dev. Biol. 198, 319329
(1998).
67. Nguyen, V. H. et al. Ventral and lateral regions of the zebrafish
gastrula, including neural crest progenitors, are established
by a bmp2b/swirl pathway of genes. Dev. Biol. 199, 93110
(1998).
68. Ikeya, M., Lee, S. M. K., Johnson, J. E., McMahon, A. P. &
Takada, S. Wnt signalling required for expansion of neural
crest and CNS progenitors. Nature 389, 966970 (1997).
69. Mayor, R., Guerrero, N. & Martinez, C. Role of FGF and
Noggin in neural crest induction. Dev. Biol. 189, 112 (1997).
70. Dorsky, R. I., Moon, R. T. & Raible, D. W. Control of neural
crest fate by the Wnt signalling pathway. Nature 396,
370373 (1998).
71. Streit, A. & Stern, C. D. Establishment and maintenance of
the border of the neural plate and in the chick: involvement of
FGF and BMP activity. Mech. Dev. 82, 5166 (1999).
72. Alvarez, I. S., Araujo, M. & Nieto, M. A. Neural induction in
whole chick embryo cultures by FGF. Dev. Biol. 199, 4254
(1998).
73. Savagner, P., Yamada, K. M. & Thiery, J. P. The zinc finger
protein Slug causes desmosome dissociation, an initial and
necessary step growth factor-induced epithelial
mesenchymal transition. J. Cell Biol. 137, 14031419 (1997).
74. Montero, J. A. et al. Role of FGFs in the control of
programmed cell death during limb development.
Development 128, 20762084 (2001).
75. Ros, M., Sefton, M. & Nieto, M. A. Slug, a zinc finger gene
previously implicated in the early patterning of the mesoderm
and the neural crest, is also involved in chick limb
development. Development 124, 18211829 (1997).
76. Buxton, P. G., Kostakopoulou, K., Brickell, P., Thorogood, P.
& Ferretti, P. Expression of the transcription factor slug
correlates with growth of the limb bud and is regulated by
FGF-4 and retinoic acid. Int. J. Dev. Biol. 41, 559568 (1997).
77. Ciruna, B. & Rossant, J. FGF signalling regulates mesoderm
cell fate specification and morphogenetic movement at the
primitive streak. Dev. Cell 1, 3749 (2001).
78. Vallin, J. et al. Cloning and characterization of three Xenopus
Slug promoters reveal direct regulation by Lef/β-catenin
signaling. J. Biol. Chem. 276, 3035030358 (2001).
79. Kwonseop, K., Zifan, L. & Hay, E. D. Direct evidence for a role
of β-catenin/LEF1 signaling pathway in induction of EMT.
Submitted.
80. Tan, C. et al. Inhibition of integrin linker kinase (ILK)
suppresses β-catenin-Lef/Tcf-dependent transcription and
expression of the E-cadherin repressor snail, in APC/
human colon carcinoma cells. Oncogene 20, 133140
(2001).
81. Willert, K. & Nusse, R. β-catenin: a key mediator of Wnt
signalling. Curr. Opin. Genet. Dev. 8, 95102 (1998).
82. Oft, M., Heider, K. & Beug, H. TGF-β signalling is necessary
for carcinoma cell invasiveness and metastasis. Curr. Biol. 8,
12431252 (1998).
83. Akhurst, R. J. & Balmain, A. Genetic events and the role of
TGF-β in epithelial tumour progression. J. Pathol. 187, 8290
(1999).
84. Isaac, A., Sargent, M. G. & Cooke, J. Control of vertebrate
leftright asymmetry by a Snail-related zinc finger gene.
Science 275, 13011304 (1997).
The first description of the involvement of Snail in
leftright asymmetry.
85. Jiang, R., Lan, Y., Norton, C. R., Sundberg, J. P. & Gridley, T.
The Slug gene is not essential for mesoderm or neural crest
development in mice. Dev. Biol. 198, 277285 (1998).
This study showed the phenotype of Slug-mutant
mice.
86. Linker, C., Bronner-Fraser, M. & Mayor, R. Relationship
between gene expression domains of Xsnail, Xslug, and
Xtwist and cell movement in the prospective neural crest of
Xenopus. Dev. Biol. 224, 215225 (2000).
87. Seher, T. C. & Leptin, M. Tribbles, a cell-cycle brake that
coordinates proliferation and morphogenesis during
Drosophila gastrulation. Curr. Biol. 10, 623629 (2000).
88. Grosshans, J. & Wieschaus, E. A genetic link between
morphogenesis and cell division during formation of the
ventral furrow in Drosophila. Cell 101, 523531 (2000).
89. Hayashi, S., Hirose, S., Metcalfe, T. & Shirras, A. D. Control of
imaginal cell development by the escargot gene of
Drosophila. Development 118, 105115 (1993).
90. Ballester, A., Frampton, J., Vilaboa, N. & Calés, C.
Heterologous expression of the transcriptional regulator
Escargot inhibits megakaryocytic endomitosis. J. Biol. Chem.
276, 4341343418 (2001).
91. Ashraf, S. I. & Ip, Y. T. The Snail protein family regulates
expression of inscutable and string, genes involved in
asymmetry and cell division in Drosophila. Development 128,
47574767 (2001).
92. Cai, Y., Chia, W. & Yang, X. A family of snail-related zinc finger
proteins regulates two distinct and parallel mechanisms that
mediate Drosophila neuroblast asymmetric divisions. EMBO
J. 20, 17041714 (2001).
The first demonstration of the involvement of Snail
family members in asymmetric cell division.
93. Capdevilla, J., Vogan, K. J., Tabin, C. J. & Izpisua-Belmonte,
J. C. Mechanisms of leftright determination in vertebrates.
Cell 101, 921 (2000).
94. Monsoro-Burq, A. H. & LeDouarin, N. BMP4 plays a role in
leftright patterning in chick embryos by maintaining Sonic
hedgehog asymmetry. Mol. Cell 7, 789799 (2001).
95. Ashraf, S. I., Hu, X., Rote, J. & Ip, Y. T. The mesoderm
determinant snail collaborates with related zinc-finger
proteins to control Drosophila neurogenesis. EMBO J. 18,
64266438 (1999).
The isolation of worniu, a third member of the snail
family in Drosophila and the demonstration of the role
of snail genes in neural differentiation.
96. Nakakura, E. K. et al. Mammalian Scratch participates in
neuronal differentiation in P19 embryonal carcinoma cells.
Mol. Brain Res. 95, 162166 (2001).
97. Fuse, N., Matakatsu, H., Taniguchi, M. & Hayashi, S. Snail-
type zinc finger proteins prevent neurogenesis in Scutoid and
transgenic animals in Drosophila. Dev. Genes Evol. 209,
573580 (1999).
98. Tanaka-Matakatsu, M., Uemura, T., Oda, H., Takeichi, M. &
Hayashi, S. Cadherin-mediated cell adhesion and motility in
Drosophila trachea regulated by the transcription factor
Escargot. Development 122, 36973705 (1996).
99. Fuse, N., Hirose, S. & Hayashi, S. Determination of wing cell
fate by the escargot and snail genes in Drosophila.
Development 122, 10591067 (1996).
100. Manzanares, M. et al. Conservation and elaboration of Hox
gene regulation during evolution of the vertebrate head.
Nature 408, 854857 (2000).
101. Holland, P. W. H., García-Fernández, J., Williams, N. A. &
Sidow, A. Gene duplications at the origin of vertebrate
development. Development S125S133 (1994).
102. Wolfe, K. Yesterday´s polyploids and the mystery of
diploidization. Nature Rev. Genet. 2, 333341 (2001).
103. Postlethwait, J. H. et al. Vertebrate genome evolution and the
zebrafish gene map. Nature Genet. 18, 345349 (1998).
104. Robinson-Rechavi, M., Marchand, O., Escriva, H. & Laudet, V.
An ancestral whole-genome duplication may not have been
responsible for the abundance of duplicated fish genes. Curr.
Biol. 11, R458R459 (2001).
105. Perl, A. K.,Wilgenbus, P., Dahl, U., Semb, H. & Christofori, G.
A causal role for E-cadherin in the transition from adenoma to
carcinoma. Nature 392, 190193 (1998).
The demonstration of a causal effect between
E-cadherin loss and epithelial tumour progression.
106. Poser, I. et al. Loss of E-cadherin expression in melanoma
cells involves upregulation of the transcriptional repressor
Snail. J. Biol. Chem. 276, 2466124666 (2001).
107. Yokoyama, K. et al. Reverse correlation of E-cadherin and
snail expression in oral squamous cell carcinoma cells in
vitro. Oral Oncol. 37, 6571 (2001).
108. Cheng, C. W. et al. Mechanisms of inactivation of E-cadherin
in breast carcinoma: modification of the two-hit hypothesis of
tumor suppressor gene. Oncogene 20, 38143823 (2001).
109. Blanco, M. J. et al. Correlation of Snail expression with
histological grade and lymph node status in breast
carcinomas. Submitted.
110. Boulay, J. L., Dennefield, C. & Alberga, A. The Drosophila
development gene snail encodes a protein with nucleic acid
binding domains. Nature 330, 395398 (1987).
The cloning of Drosophila snail.
111. Whiteley, M., Noguchi, P. D., Sensaburgh, S. M., Odenwald,
W. F. & Kassis, J. A. The Drosophila gene escargot encodes
a zinc finger motif found in snail-related genes. Mech. Dev.
36, 117127 (1992).
112. Smith, S., Metcalfe, J. A. & Elgar, G. Identification and
analysis of two snail genes in the pufferfish (Fugu rubripes)
and mapping of human SNA to 20q. Gene 247, 119128
(2000).
113. Sargent, M. G. & Bennet, M. F. Identification in Xenopus of a
structural homologue of the Drosophila gene snail.
Development 109, 963973 (1990).
The isolation of the first vertebrate Snail homologue.
114. Mayor, R., Morgan, R. & Sargent M. G. Induction of the
prospective neural crest of Xenopus. Development 121,
767777 (1995).
115. Paznekas, W. A., Okajima, K., Schertzer, M., Wood, S. &
Jabs, E. W. Genomic organization, expression, and
chromosome location of the human SNAIL gene (SNAI1)and
a related processed pseudogene (SNAI1P). Genomics 62,
4249 (1999).
116. Twigg, S. R. F. & Wilkie, A. O. M. Characterisation of the
human snail (SNAI1) gene and exclusion as a major disease
gene in craniosynostosis. Hum. Genet. 105, 320326 (1999).
117. Rhim, H., Savagner, P., Thibaudeau, G., Thiery, J. P. & Pavan,
W. P. Localization of a neural crest transcription factor, Slug,
to mouse chromosome 16 and human chromosome 8.
Mamm. Genome 8, 872873 (1997).
Acknowledgements
I am very grateful to people in the lab for their work along the years
and for their encouraging discussions. Work in the lab is, at pre-
sent, supported by grants from the Spanish Ministries of Science
and Technology (DGESIC) and Health (FIS), and from the Local
Government (Comunidad Autónoma de Madrid).
Online links
DATABASES
The following terms in this article are linked online to:
FlyBase: http://flybase.bio.indiana.edu/
CtBP | deadpan | RhoGEF2 | Escargot | Fog | rhomboid | scratch |
single-minded | String | snail | Tribbles | worniu
LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink
BCL-X
L
| ILK
Swiss-Prot: http://www.expasy.ch/
β-catenin | cytokeratin-18 | desmoplakin | E-cadherin | FGFR1 |
fibronectin | Gfi1 | HLF | Lef-1 | Muc-1 | Nodal | PTH(rP) | RhoB |
SIP1 | Slug | Smuc | snail1 | snail2 | TGF-β1 | TGF-β2 | vimentin
WormBase: http://www.wormbase.org/
ces-1 | CES-2 | CED-3 | CED-4 | ced-9 | EGL-1
Access to this interactive links box is free online.
... It acted as a boundary repressor to regulate mesodermal differentiation by down-regulating ectodermal gene expression within the mesoderm [6]. Its conserved functions were also presented in insects, urochordates, and vertebrates [7,8]. There are three Snail family proteins in vertebrates, namely Snail1 (Snail), Snail2 (Slug), and Snail3 (Smuc). ...
... It has been shown that the vertebrate Snail family members Snail and Slug are involved in the regulation of the EMT processes by down-regulating mesenchymal markers (Fibronectin, Vimentin) and up-regulating cell junction-associated proteins (E-cadherin, Claudin, and Plakoglobin) [10]. However, only one Snail protein was identified in Ciona without a SNAG box [7], and its function remains to be elucidated. ...
... Snail, as an EMT-related transcription repressor, is required for mesoderm and neural crest formation during early embryonic development [7,8]. Our previous work detecting temporal expression of Ci-Snail by RT-qPCR showed that it presented high expression during gastrulation (16 • C, 10 hpf) but decreased expression during the early tailbud stage (16 • C, 14 hpf); however, elevated expression again at late tailbud stage, accordingly the MET initiation stage (16 • C, 18 hpf) ( Figure S1). ...
Article
Full-text available
Mesenchymal-epithelial transition (MET) is a widely spread and evolutionarily conserved process across species during development. In Ciona embryogenesis, the notochord cells undergo the transition from the non-polarized mesenchymal state into the polarized endothelial-like state to initiate the lumen formation between adjacent cells. Based on previously screened MET-related transcription factors by ATAC-seq and Smart-Seq of notochord cells, Ciona robusta Snail (Ci-Snail) was selected for its high-level expression during this period. Our current knockout results demonstrated that Ci-Snail was required for notochord cell MET. Importantly, overexpression of the transcription factor Brachyury in notochord cells resulted in a similar phenotype with failure of lumen formation and MET. More interestingly, expression of Ci-Snail in the notochord cells at the late tailbud stage could partially rescue the MET defect caused by Brachyury-overexpression. These results indicated an inverse relationship between Ci-Snail and Brachyury during notochord cell MET, which was verified by RT-qPCR analysis. Moreover, the overexpression of Ci-Snail could significantly inhibit the transcription of Brachyury, and the CUT&Tag-qPCR analysis demonstrated that Ci-Snail is directly bound to the upstream region of Brachyury. In summary, we revealed that Ci-Snail promoted the notochord cell MET and was essential for lumen formation via transcriptionally repressing Brachyury.
... On the other hand, the Index case expressed high mRNA levels of mesenchymal markers, such as MMP3, FN1 and Vimentin ( Figure 2C). We did not find significant differences in SNAI1, whose functions are associated with development processes, neural differentiations and epitheliummesenchymal transition [15]. The replacement of usual epithelial markers with mesenchymal markers, such as Vimentin and Fibronectin, determines these changes [15,16]. ...
... We did not find significant differences in SNAI1, whose functions are associated with development processes, neural differentiations and epitheliummesenchymal transition [15]. The replacement of usual epithelial markers with mesenchymal markers, such as Vimentin and Fibronectin, determines these changes [15,16]. Figure 2A and 2C confirm a decrease in endothelial markers and an increase in mesenchymal markers, supporting our initial idea that an EndMT is occurring. ...
Preprint
Full-text available
Pulmonary arteriovenous malformations (PAVMs) are vascular anomalies resulting in abnormal connection between pulmonary arteries and veins. In 80% of cases, PAVMs are present from birth, but clinical manifestations are rarely seen in childhood. These congenital malformations are typically associated with Hereditary Hemorrhagic Telangiectasia (HHT), a rare disease that affects 1 in 5,000/8,000 individuals. HHT disease is frequently caused by mutations in genes involved in the TGF-β pathway. However, approximately 15% of patients do not have a genetic diagnosis and, among the genetically diagnosed, more than 33% do not meet the Curaçao Criteria. This makes clinical diagnosis even more challenging in the pediatric age group. Here, we introduce an 8 years old patient bearing a severe phenotype of multiple diffuse PAVMs caused by an unknown mutation which ended in lung transplantation. Phenotypically, the case of study follows a molecular pattern HHT-like. Therefore, molecular biology and cellular functional analysis has been performed in primary endothelial cells (ECs) isolated from the explanted lung. The findings revealed a loss of functionality in lung endothelial tissue and a stimulation of endothelial to mesenchymal transition. Understanding the molecular basis of this transition could potentially offer new therapeutic strategies to delay lung transplantation in severe cases.
... This gene superfamily exhibits functional redundancy in both vertebrates and insects, primarily in the development of the nervous system. For instance, in chickens, snai2, rather than snai1, is expressed in the premigratory neural crest and the primitive streak (Nieto, 2002), snai2 is involved in neural crest formation; its overexpression leads to an increase in neural crest production. This phenotype is mimicked by ectopic expression of snai1 (del Barrio & Nieto, 2002). ...
Article
Full-text available
The Snail superfamily of transcription factors plays a crucial role in metazoan development; one of the most important vertebrate members of this family is Snai1 which is orthologous to the Drosophila melanogaster esg gene. This review offers a comprehensive examination of the roles of the esg gene in Drosophila development, covering its expression pattern and downstream targets, and draws parallels between the vertebrate Snai1 family proteins on controlling the epithelial‐to‐mesenchymal transition and esg . This gene regulates stemness, ploidy, and pluripontency. esg is expressed in various tissues during development, including the gut, imaginal discs, and neuroblasts. The functions of the esg include the suppression of differentiation in intestinal stem cells and the preservation of diploidy in imaginal cells. In the nervous system development, esg expression also inhibits neuroblast differentiation, thus regulating the number of neurons and the moment in development of neuronal differentiation. Loss of esg function results in diverse developmental defects, including defects in intestinal stem cell maintenance and differentiation, and alters imaginal disc and nervous system development. Expression levels of esg also play a role in regulating longevity and metabolism in adult stages. This review provides an overview of the current understanding of esg's developmental role, emphasizing cellular and tissue effects that arise from its loss of function. The insights gained may contribute to a better understanding of evolutionary conserved developmental mechanisms and certain metabolic diseases.
... Recent research has elucidated intricate regulatory networks involving Snail and Slug in EMT progression. [78][79][80][81] Our choice of these genes was grounded in their well-established roles and interactions, providing a comprehensive view of the EMT process and its modulation in response to Taxifolin and EPI. ...
Article
Full-text available
Background: One of the most significant characteristics of cancer is epithelial–mesenchymal transition and research on the relationship between phenolic compounds and anticancer medications and epithelial–mesenchymal transition is widespread. Methods: In order to investigate the potential effects of Taxifolin on enhancing the effectiveness of Epirubicin in treating breast cancer, specifically in 4T1 cells and an allograft BALB/c model, the effects of Taxifolin and Epirubicin, both individually and in combination, were examined. Cell viability assays and cytotoxicity assays in 4T1 cells were performed. In addition, 4T1 cells were implanted into female BALB/c mice to conduct in vivo studies and evaluate the therapeutic efficacy of Taxifolin and Epirubicin alone or in combination. Tumor volumes and histological analysis were also assessed in mice. To further understand the mechanisms involved, we examined the messenger RNA and protein levels of epithelial–mesenchymal transition-related genes, as well as active Caspase-3/7 levels, using quantitative real-time polymerase chain reaction, western blot, and enzyme-linked immunosorbent assays, respectively. Results: In vitro results demonstrated that the coadministration of Taxifolin and Epirubicin reduced cell viability and cytotoxicity in 4T1 cell lines. In vivo, coadministration of Taxifolin and Epirubicin suppressed tumor growth in BALB/c mice with 4T1 breast cancer cells. Additionally, this combination treatment significantly increased the levels of active caspase-3/7 and downregulated the messenger RNA and protein levels of N-cadherin, β-catenin, vimentin, snail, and slug, but upregulated the E-cadherin gene. It significantly decreased the messenger RNA levels of the Zeb1 and Zeb2 genes. Conclusion: The in vitro and in vivo results of our study indicate that the concurrent use of Epirubicin with Taxifolin has supportive effects on breast cancer treatment.
... Despite, Ca Ski RT_A subclones demonstrated an increase in the expression of E-CADHERIN manifesting a loss of the typical heterogeneous morphology of epithelial cells. Besides, Ca Ski RT_A exhibited decreased levels of expression of VIMENTIN, a type III intermediate filament expressed in the mesenchymal cells and upregulated during cancer metastasis [102], as well as of the transcription factors Snai1 (Snail) and Snai2 (Slug) (but not for all subclones), master regulatory factors for organogenesis and wound healing, tightly involved in EMT of cancer cells [110][111][112]. These data indicated that Ca Ski RT_A subclones acquired features of the hybrid epithelial/mesenchymal (E/M) phenotype where cells simultaneously demonstrate the epithelial traits of cell-to-cell adhesion and mesenchymal characteristics of migration and invasion [113]. ...
Preprint
Full-text available
High incidence of epithelial malignancies in HIV-1 infection is associated with co-infection with oncogenic viruses, such as high-risk human papillomaviruses (HR HPVs), most often HPV16. A collaboration between HIV-1 and HR HPVs in the malignant transformation of epithelial cells exposed to both viruses have long been anticipated. Here, we delineated the effects on in vitro and in vivo properties of HPV16-infected cervical cancer cells of HIV-1 reverse transcriptase. Epidermoid carcinoma Ca Ski cells were made to express RT of HIV-1 clade A (RT_A) or Green Fluorescence Protein (GFP) by lentiviral transduction, subclones with one or six copies of RT_A and six copies of GFP DNA were generated. Expression by subclones of GFP was assessed by flow cytometry, and of RT_A, by Western blotting. Cells were assessed for the levels of mRNA of the RT_A, E6 and its isoforms, and cellular factors characterizing oxidative stress, and EMT by the real-time PCR. Parameters of glycolysis and mitochondrial respiration were assessed by Seahorse technology. Subclones were surveyed for the changes in cell cycle, doubling time, migration capacity, clonogenic activity and for the capacity to form tumors in nude mice. Ca Ski RT_A produced 20-55 fg of RT_A per cell. Expression of RT_A caused a proportional increase in the expression of E6*I isoform. In Ca Ski, RT_A suppressed mitochondrial respiration and oxygen consumption, and increased glycolysis. Compared to parental cells and GFP control, transcription in Ca Ski RT_A of epithelial (E-CADHERIN) state marker was increased, and of mesenchymal (VIMENTIN) decreased, indicating acquisition by cells of the hybrid epithelial/mesenchymal (E/M) phenotype. While Ca Ski GFP and Ca Ski RT_A with one gene insert had reduced migration rate, decreased clonogenic activity in vitro, and diminished capacity to form tumors in nude mice, increase in RT_A copicity/expression mitigated these effects. Altogether, expression of HIV-1 RT_A gave Ca Ski cells the plasticity required to overcome negative effects of lentiviral transduction and potentially increased their tumorigenicity.
Article
Full-text available
Tumor biomarkers, the substances which are produced by tumors or the body’s responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Article
Full-text available
Breast cancer is one of the prevailing cancers globally, with a high mortality rate. Metastatic breast cancer (MBC) is an advanced stage of cancer, characterised by a highly nonlinear, heterogeneous process involving numerous singling pathways and regulatory interactions. Epithelial–mesenchymal transition (EMT) emerges as a key mechanism exploited by cancer cells. Transforming Growth Factor-β (TGFβ)-dependent signalling is attributed to promote EMT in advanced stages of breast cancer. A comprehensive regulatory map of TGFβ induced EMT was developed through an extensive literature survey. The network assembled comprises of 312 distinct species (proteins, genes, RNAs, complexes), and 426 reactions (state transitions, nuclear translocations, complex associations, and dissociations). The map was developed by following Systems Biology Graphical Notation (SBGN) using Cell Designer and made publicly available using MINERVA ( http://35.174.227.105:8080/minerva/?id=Metastatic_Breast_Cancer_1 ). While the complete molecular mechanism of MBC is still not known, the map captures the elaborate signalling interplay of TGFβ induced EMT-promoting MBC. Subsequently, the disease map assembled was translated into a Boolean model utilising CaSQ and analysed using Cell Collective. Simulations of these have captured the known experimental outcomes of TGFβ induced EMT in MBC. Hub regulators of the assembled map were identified, and their transcriptome-based analysis confirmed their role in cancer metastasis. Elaborate analysis of this map may help in gaining additional insights into the development and progression of metastatic breast cancer.
Article
Full-text available
NUT carcinoma (NC) is an aggressive cancer with no effective treatment. About 70% of NUT carcinoma is associated with chromosome translocation events that lead to the formation of a BRD4::NUTM1 fusion gene. Because the BRD4::NUTM1 gene is unequivocally cytotoxic when ectopically expressed in cell lines, questions remain on whether the fusion gene can initiate NC. Here, we report the first genetically engineered mouse model for NUT carcinoma that recapitulates the human t(15;19) chromosome translocation in mice. We demonstrated that the mouse t(2;17) syntenic chromosome translocation, forming the Brd4::Nutm1 fusion gene, could induce aggressive carcinomas in mice. The tumors present histopathological and molecular features similar to human NC, with enrichment of undifferentiated cells. Similar to the reports of human NC incidence, Brd4::Nutm1 can induce NC from a broad range of tissues with a strong phenotypical variability. The consistent induction of poorly differentiated carcinoma demonstrated a strong reprogramming activity of BRD4::NUTM1. The new mouse model provided a critical preclinical model for NC that will lead to better understanding and therapy development for NC.
Article
Full-text available
Patient‐Derived Organoids (PDO) and Xenografts (PDX) are the current gold standards for patient‐derived models of cancer (PDMC). Nevertheless, how patient tumor cells evolve in these models and the impact on drug response remains unclear. Herein, the transcriptomic and chromatin accessibility landscapes of matched colorectal cancer (CRC) PDO, PDX, PDO‐derived PDX (PDOX), and original patient tumors (PT) are compared. Two major remodeling axes are discovered. The first axis delineates PDMC from PT, and the second axis distinguishes PDX and PDO. PDOX are more similar to PDX than PDO, indicating the growth environment is a driving force for chromatin adaptation. Transcription factors (TF) that differentially bind to open chromatins between matched PDO and PDOX are identified. Among them, KLF14 and EGR2 footprints are enriched in PDOX relative to matched PDO, and silencing of KLF14 or EGR2 promoted tumor growth. Furthermore, EPHA4, a shared downstream target gene of KLF14 and EGR2, altered tumor sensitivity to MEK inhibitor treatment. Altogether, patient‐derived CRC cells undergo both common and distinct chromatin remodeling in PDO and PDX/PDOX, driven largely by their respective microenvironments, which results in differences in growth and drug sensitivity and needs to be taken into consideration when interpreting their ability to predict clinical outcome.
Article
Full-text available
UPP1, a crucial pyrimidine metabolism-related enzyme, catalyzes the reversible phosphorylation of uridine to uracil and ribose-1-phosphate. However, the effects of UPP1 in bladder cancer (BLCA) have not been elucidated. AKT, which is activated mainly through dual phosphorylation (Thr308 and Ser473), promotes tumorigenesis by phosphorylating downstream substrates. This study demonstrated that UPP1 promotes BLCA cell proliferation, migration, invasion, and gemcitabine resistance by activating the AKT signaling pathway in vitro and in vivo. Additionally, UPP1 promoted AKT activation by facilitating the binding of AKT to PDK1 and PDK2 and the recruitment of phosphatidylinositol 3,4,5-triphosphate to AKT. Moreover, the beneficial effects of UPP1 on BLCA tumorigenesis were mitigated upon UPP1 mutation with Arg94 or MK2206 treatment (AKT-specific inhibitor). AKT overexpression or SC79 (AKT-specific activator) treatment restored tumor malignancy and drug resistance. Thus, this study revealed that UPP1 is a crucial oncogene and a potential therapeutic target for BLCA and that UPP1 activates the AKT signaling pathway and enhances tumorigenesis and drug resistance to gemcitabine.
Article
Full-text available
Mutations in the escargot (esg) locus, which codes for a zinc-finger-containing protein with similarity to the product of the snail gene, cause a variety of defects in adult structures such as loss of abdominal cuticle and malformation of the wings and legs. esg RNA is expressed in wing, haltere, leg and genital imaginal discs and in abdominal histoblast nests in the embryo. Expression in imaginal tissues is also found in third instar larvae. In esg mutant larvae, normally diploid abdominal histoblasts replicate their DNA without cell division and become similar in appearance to the polytene larval epidermal cells. A similar phenotype was also found in imaginal discs of larvae mutant for both esg and the Drosophila raf gene. These results suggest that one of the normal functions of esg may be the maintenance of diploidy in imaginal cells.
Article
We identified two cadherins, c-cad6B and c-cad7, expressed by neural crest cells at their premigratory and migratory stages, respectively, in chicken embryos. cDNA transfection experiments showed that both were homophilic adhesion molecules, endowing cells with specific adhesiveness. During development, c-cad6B appeared in the neural fold, localizing at the future neural crest area. This expression was maintained during neural tube closure, but disappeared after neural crest cells had left the neural tube, suggesting its role in neural fold fusion and/or in the formation and maintenance of the presumptive neural crest domain in the neural plate/tube. Crest cells emerging from the neural tube lost c-cad6B, and a subpopulation of them began to express c-cad7. This subpopulation-specific expression of c-cad7 persisted during their migration. The migrating c-cad7-positive cells clustered together, and eventually populated restricted regions including the dorsal and ventral roots but very little ganglia. The latter was populated with N-cadherin-positive crest cells. Migrating neural crest cells expressed alpha- and beta-catenin at cell-cell contacts, indicating that their cadherins are functioning. These results suggest that the migrating crest cells are grouped into subpopulations expressing different cadherins. The cadherin-mediated specific interaction between crest cells likely plays a role in intercellular signaling between homotypic cells as well as in sorting of heterotypic cells.
Article
The mouse skin model of chemical carcinogenesis has been very well characterized with respect to epigenetic changes, which occur during tumour cell initiation, promotion and progression. The use of transgenic and gene knock‐out mice has contributed greatly to knowledge in this area. The H‐ras genetic locus has been shown to undergo multiple genetic changes, including mutagenic activation, amplification of the mutant gene, and loss of the normal allele. These different genetic events lead to thresholds of ras activity which contribute to different stages along the pathway to neoplasia. The genetic and epigenetic events which lead to tumour invasion and metastasis have been less well characterized than studies on tumour initiation and promotion, despite the fact that it is metastases which ultimately kill the animal/patient. In the mouse skin model, loss of p53 contributes to malignant conversion. Gene deletion of the INK4 locus is associated with transformation to a highly invasive spindle cell tumor phenotype. This spindle cell transformation can also be induced in vitro or in vivo by TGFβ 1, possible by synergizing with mutant H‐ras. TGFβ can have both positive and negative effects on tumourigenesis, acting early as a tumour suppresser, but later as a stimulator of tumour invasion. It is this latter effect which may be clinically more significant, since many human tumours overexpress TGFβ, yet the majority still retain the intracellular signaling systems necessary for the cell to respond to this growth factor. Copyright © 1999 John Wiley & Sons, Ltd.
Article
In a search for embryonic lethal mutants on the second chromosome ofDrosophila melanogaster, 5764 balanced lines isogenic for an ethyl methane sulfonate (EMS)-treatedcn bw sp chromosome were established. Of these lines, 4217 carried one or more newly induced lethal mutations corresponding to a total of 7600 lethal hits. Eggs were collected from lethal-bearing lines and unhatched embryos from the lines in which 25% or more of the embryos did not hatch (2843 lines) were dechorionated, fixed, cleared and scored under the compound microscope for abnormalities of the larval cuticle. A total of 272 mutants were isolated with phenotypes unequivocally distinguishable from wild-type embryos on the basis of the cuticular pattern. In complementation tests performed between mutants with similar phenotype, 48 loci were identified by more than one allele, the average being 5.4 alleles per locus. Complementation of all other mutants was shown by 13 mutants. Members of the complementation groups were mapped by recombination analysis. No clustering of loci with similar phenotypes was apparent. From the distribution of the allele frequencies and the rate of discovery of new loci, it was estimated that the 61 loci represent the majority of embryonic lethal loci on the second chromosome yielding phenotypes recognizable in the larval cuticle.
Article
Snail family proteins are zinc finger transcriptional regulators first identified in Drosophila which play critical roles in cell fate determination. We identified a novel Snail-related gene from murine skeletal muscle cells designated Smuc. Northern blot analysis showed that Smuc was highly expressed in skeletal muscle and thymus. Smuc contains five putative DNA-binding zinc finger domains in its C-terminal half. In electrophoretic mobility shift assays, recombinant zinc finger domains of Smuc specifically bound to CAGGTG and CACCTG E-box motifs (CANNTG). Because basic helix–loop–helix transcription factors (bHLH) bind to the same E-box sequences, we examined whether Smuc competes with the myogenic bHLH factor MyoD for DNA binding. Smuc inhibited the binding of a MyoD–E12 complex to the CACCTG E-box sequence in a dose-dependent manner and suppressed the transcriptional activity of MyoD–E12. When heterologously targeted to the thymidine kinase promoter as fusion proteins with the GAL4 DNA-binding domain, the non-zinc finger domain of Smuc acted as a transcriptional repressor. Furthermore, overexpression of Smuc in myoblasts repressed transactivation of muscle differentiation marker Troponin T. Thus, Smuc might regulate bHLH transcription factors by zinc finger domains competing for E-box binding, and non-zinc finger repressor domains might also confer transcriptional repression to control differentiation processes.
Article
Transcripts of a newly discovered gene calledsnail2,encoding a zinc finger protein of the Snail family, first appear in rows of cephalic mesendodermal cells in gastrulating zebrafish embryos. At the end of gastrulation,snail2RNA accumulates in a domain of ectodermal cells that mark the border between the epidermal epithelium and the neural plate and includes precursors of the neural crest. During somitogenesis,snail2expression becomes restricted to neural crest.snail2is thus one of the earliest genes yet known to be specifically expressed in neural crest in zebrafish embryos. Sincesnail2is expressed in mesendoderm, a tissue layer whose convergence in the trunk is known to be altered in embryos homozygous for thespadetailmutation, we examinedsnail2expression inspadetailembryos. In these mutants, the number of cephalic mesendodermal cells expressingsnail2is strongly reduced and the distribution of cells containingsnail2andno tailtranscripts in the axial mesoderm is much broader than normal. Moreover, the embryos are shorter than normal at the end of gastrulation. This shows that, in addition to the failure of paraxial mesoderm to converge normally in the trunk during gastrulation,spadetailalso affects the elongation of the embryo and the convergence of axial and lateral mesendoderm in both trunk and head.