You are on page 1of 19

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/24216628

The Role of the Visceral Mesoderm in the Development of the Gastrointestinal


Tract

Article  in  Gastroenterology · April 2009


DOI: 10.1053/j.gastro.2009.03.001 · Source: PubMed

CITATIONS READS
94 871

3 authors, including:

Valerie Mclin Susan J. Henning


University of Geneva University of North Carolina at Chapel Hill
180 PUBLICATIONS   2,728 CITATIONS    185 PUBLICATIONS   6,157 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Chronic hepatic encephalopathy View project

Intestinal mucosal homeostasis View project

All content following this page was uploaded by Susan J. Henning on 05 February 2018.

The user has requested enhancement of the downloaded file.


GASTROENTEROLOGY 2009;136:2074 –2091
GASTROENTEROLOGY
BASIC AND CLINICAL

REVIEWS IN BASIC AND CLINICAL


REVIEWS IN

GASTROENTEROLOGY
John P. Lynch and David C. Metz, Section Editors

The Role of the Visceral Mesoderm in the Development of the


Gastrointestinal Tract

VALÉRIE A. MCLIN,* SUSAN J. HENNING,‡ and MILAN JAMRICH§,储


*Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, §Department of Cellular and Molecular Biology, and 储Department of Molecular and
Human Genetics, Baylor College of Medicine, Houston, Texas; and ‡Departments of Medicine and Cell and Molecular Physiology, University of North Carolina Chapel
Hill, Chapel Hill, North Carolina

The gastrointestinal (GI) tract forms from the en- served from invertebrates to vertebrates, including the
doderm (which gives rise to the epithelium) and role of the visceral mesoderm (VM). Understanding the
the mesoderm (which develops into the smooth mus- role of the VM in development may help orient cell-based
cle layer, the mesenchyme, and numerous other cell therapy research.
types). Much of what is known of GI development has
been learned from studies of the endoderm and its
derivatives, because of the importance of epithelial Overview of GI Development
biology in understanding and treating human dis- Development of the vertebrate GI tract, and of the
eases. Although the necessity of epithelial-mesenchy- VM in particular, is conserved across species. In very
mal cross talk for GI development is uncontested, the broad terms, it follows this sequence of events: gastrula-
role of the mesoderm remains comparatively less well tion, formation of the primitive gut tube from the
understood. The transformation of the visceral me- endoderm, and apposition of the inner leaflet of the
soderm during development is remarkable; it differ- lateral plate mesoderm against the endoderm.1 This inner
entiates from a very thin layer of cells into a complex leaflet eventually circles the gut to become the VM (Fig-
tissue comprising smooth muscle cells, myofibro- ure 1). The outer leaflet becomes the somatic muscula-
blasts, neurons, immune cells, endothelial cells, lym- ture.1 During GI morphogenesis, endoderm and meso-
phatics, and extracellular matrix molecules, all con- derm undergo extensive regionalization, elongation, and
tributing to the form and function of the digestive coiling. Concurrently, neural crest cells populate the in-
system. Understanding the molecular processes that testine to form the early enteric nervous system (ENS).
govern the development of these cell types and eluci- This period of rapid growth is characterized by regional
dating their respective contribution to GI patterning signals that are largely derived from the mesoderm and
could offer insight into the mechanisms that regulate pattern the future intestinal domains along 4 axes: ante-
cell fate decisions in the intestine, which has the rior-posterior, dorsoventral, left-right, and radial. The
unique property of rapid cell renewal for the mainte- mesoderm sends (or receives) instructive or permissive
nance of epithelial integrity. In reviewing evidence signals to the adjacent endoderm; the instructive signals
from both mammalian and nonmammalian models, are sufficient to direct cell fate in the target tissue,
we reveal the important role of the visceral mesoderm whereas permissive signals enable competent tissue to
in the ontogeny of the GI tract. activate a previously “primed” genetic program.1 The
most likely scenario in GI patterning is that neither germ
layer holds all the information necessary for GI develop-

I nherited or acquired abnormalities of the gastrointes-


tinal (GI) tract cause many chronic diseases in children
and adults that often require costly, long-term medical
Abbreviations used in this paper: BMP, bone morphogenetic protein;
E, embryonic day; ECM, extracellular matrix; ENS, enteric nervous
support and organ transplantation. Cell-based therapies system; FGF, fibroblast growth factor; GC, glucocorticoid; GI, gastroin-
are an attractive alternative to solid organ transplanta- testinal; Hh, Hedgehog; IGF, insulin-like growth factor; ISEMF, intesti-
tion, but their progress requires a better understanding nal subepithelial myofibroblast; PDGF, platelet-derived growth factor;
SOCS, suppressor-of-cytokine signaling; VM, visceral mesoderm.
of the processes that govern cell fate decisions, cell pro- © 2009 by the AGA Institute
liferation, and tissue differentiation during GI develop- 0016-5085/09/$36.00
ment. Several aspects of GI development are well con- doi:10.1053/j.gastro.2009.03.001
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2075

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
DORSAL

SM N e
S S
VM

BMP4
Endoderm

Hh
FoxF1

Iro

VENTRAL
Figure 1. Early development of the lateral plate mesoderm. Diagram of a cross section through a Xenopus neurula. Dorsal is to the top. The visceral
mesoderm (VM, orange) and somatic mesoderm (SM, brown) are 2 leaflets that are in continuity with the somitic (S) mesoderm at the lateral plate.
They are situated between the endoderm (yellow) and the ectoderm (e). Endodermal Hh signals produced by the future gut endoderm induce BMP-4
expression in the VM. The ensuing molecular decisions in the VM are shown; BMP-4 activates expression of the transcription factor FoxF1. One of
the early actions of FoxF1 is the inhibition of Iroquois (Iro) in the VM, thereby inhibiting the somatic mesoderm program in cells destined to become
VM. FoxF1 also induces BMP4 expression. N, notochord; S, somitic mesoderm. Modified and reprinted with permission from El-Hodiri et al.167

ment, but rather that the relative contribution of each Following morphogenesis, the characteristic GI cell
tissue varies during ontogeny, with their relative plastic- types emerge from the undifferentiated endoderm and
ity diminishing as development proceeds.2 Although the the VM differentiates into its specialized components.
mesoderm has an important role in early GI tract mor- This phase, which begins around embryonic day (E) 15 in
phogenesis, its formation requires signals from the mice and continues postnatally, occurs during metamor-
endoderm mediated in part by the Hedgehog (Hh) family phosis in the frog and begins toward the end of the first
of signaling molecules3; this pathway is conserved among trimester in humans.16 It coincides with the formation of
species.4 – 8 the GI tract’s fifth axis, the villus-crypt axis. This axis is
Differential regional responses to the Hh signals along the functional unit of the intestine and is characterized
the craniocaudal axis lead to the formation of the fo- by finger-like evaginations into the lumen, comprising a
regut, midgut, and hindgut domains.3,5,9 –11 The foregut mesenchymal stalk and an epithelial cover composed of
gives rise to the esophagus, stomach, duodenum, liver the precursors to the different epithelial lineages. The
and bile ducts, pancreas, lung, and thyroid. The midgut is mesenchyme contains (intestinal) subepithelial myofi-
the main precursor of the intestine that is distal to the broblasts (ISEMFs), both in the stalk and adjacent to the
entry of the common bile duct and extends to the prox- intervillus space, which are important regulators of mes-
imal transverse colon; this large domain includes the enchymal signaling. In addition, the mesenchyme, also
jejunum, ileum, cecum, and ascending colon. Finally, the derived from the VM, forms the lamina propria, together
hindgut evolves into the distal transverse, descending, with blood vessels, lymphatics, resident immune cells,
and rectosigmoid segments of the colon.1,12 The contri- and the muscularis propria (Figure 2). In the fully mature
bution of mesoderm components to endoderm pattern- GI tract, mesodermal derivatives outnumber endodermal
ing is well understood for development of the liver and derivatives (Figure 2). A brief overview of the develop-
pancreas. The development of these 2 organs, which also ment of these different, mesodermally derived compo-
depends on instructive and permissive signals from the nents is warranted before examining the molecular net-
adjacent mesoderm, is the subject of recent reviews.13–15 work governing VM development.
2076 MCLIN ET AL GASTROENTEROLOGY Vol. 136, No. 7
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

Figure 2. Schematic section through the adult GI tract. Each quadrant represents a different segment of the GI tract, from the stomach to the colon;
mesenchymal structures are shown in detail. The following structures are conserved along the craniocaudal axis: the lamina propria mesenchyme, the
muscularis mucosa, mesenchymal blood vessels, the ISEMFs, the Meissner’s plexus, the longitudinal inner and circular outer layers of the muscularis
propria, the Auerbach plexus, and the serosa. These structures are represented in all quadrants. The stomach (left upper quadrant) is characterized by an
additional oblique inner layer of the muscularis propria. Its epithelium is characterized by deep, bifid glands (g) that fulfill the secretory and endocrine functions
of the stomach. In the duodenum (left lower quadrant), the submucosa is recognizable by its abundance of Brunner’s glands, which are epithelial in origin.
Consistent with the rest of the small intestine, the duodenal epithelium is characterized by the presence of crypts and villi. The other 2 components of the
small intestine are the jejunum and ileum (right lower quadrant). The ileum is characterized by the presence of Peyer’s patches, which are large lymphoid
aggregates in the submucosa. In the jejunum and the colon, the lymphoid aggregates are located in the mucosal tunic rather than in the submucosa.
ISEMFs are adjacent to the epithelium lining crypts and villi. Finally, in the colon (right upper quadrant), the epithelium is characterized by straight glands (g)
but does not include villi or crypts. The characteristic feature of the colon mesenchyme is the thinner, circular outer layer of the muscularis propria. Modified
and reprinted with permission from T. Caceci, DVM (Virginia-Maryland Regional College of Veterinary Medicine).

ISEMFs reside within the mesenchymal layer of the intestine at


As their name suggests, ISEMFs are cells that are the base of the crypts, next to the muscularis, starting
located just below the epithelium of both villi and cry- around week 21 of human gestation.18 In mouse em-
pts throughout the length of the intestine (Figure 2). bryos, ISEMFs have been described as early as E18.5.19
ISEMFs are characterized by ␣–smooth muscle actin ex- Similar to epithelial cells, ISEMFs differentiate as they
pression and are organized in a syncytium between the migrate up the crypt to villus axis.20 There is increasing
epithelium and the muscularis mucosa, where they con- evidence that ISEMFs are major players in GI development.
tribute to extracellular matrix (ECM) and basal mem-
brane composition.17 Although it is uncertain whether The Development of the GI Smooth Muscle
ISEMFs are derived from the neural crest, mesodermally The visceral smooth muscle, derived from the VM,
derived fibroblasts, or smooth muscle cells, they clearly is a very large component of the intestine. Its function in
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2077

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
the adult intestine is to confer shape and motility to this
hollow organ and allow for the advancement of its lumi-
nal contents. The muscle layers of the intestine include
the circular inner layer and the longitudinal external
layer. In the stomach, there is an additional oblique layer
(Figure 2). Together, the smooth muscle layers are called
the muscularis propria. Mammals have an additional
muscularis mucosa that separates the epithelium from
the underlying smooth muscle layers and promotes epi-
thelial movement21 (Figure 2). In humans, disorders of
enteric myocytes have been described in children with
motility syndromes22; a few reports have associated hyp-
oplasia of the muscularis propria with necrotizing en-
terocolitis or intestinal perforation in infants.23,24
Little is known about the role of the developing
smooth muscle layer during ontogeny. The early muscle
progenitors differentiate from the loose mesenchyme Figure 3. Early specification of mesodermal tissues. Diagram repre-
senting the action of BMP-4 as a ventralizing morphogen. In most
that surrounds the primitive gut. The earliest precursor species, BMP signaling in combination with other pathways patterns
to both types of enteric smooth muscle cells is a common the early mesoderm into different cell types.168 –170 In Xenopus, ze-
progenitor called the smooth muscle myoblast, which is brafish, and other species, BMP-4 acts as a mesodermal morphogen
characterized by the expression of ␣–smooth muscle ac- that specifies different mesodermal derivatives along the dorsoventral
tin, as opposed to the more differentiated smooth mus- axis: blood, muscle, kidney, and notochord.170 –174 According to its
concentration at a given time and place, BMP-4 confers a distinct
cle myocytes, which express ␣– and ␥–smooth muscle dorsoventral identity to different parts of the mesoderm.170,175 In Xeno-
actins.25,26 Formation of the smooth muscle layer pro- pus and zebrafish, the Vent homeobox-containing genes act down-
gresses in a craniocaudal fashion, starting in the esoph- stream of BMP-4 to convert the signaling gradient into distinct cellular
agus at E1125 in mice and 50 hours postfertilization in responses.176 –178 The essential VM transcription factor FoxF1 is also
zebrafish.27 This first layer of smooth muscle myoblasts downstream from BMP-4.36 Although the exact “dose” of BMP-4 that
regulates the specification and differentiation of lateral plate mesoderm
differentiates from undifferentiated mesenchyme to give has not been established, integration of combinatorial regulatory signals
rise to the circular inner layer. The second, longitudinal, is an important paradigm that is repeated throughout development. The
external layer appears in the mesenchyme approximately Vent pathway of ventral mesoderm specification has been confirmed in
24 –28 hours later.25,26 Finally, in mammals, the muscu- zebrafish and amphioxus,170,179 but mammalian studies are still
laris mucosa forms subjacent to the epithelium. In each pending.
layer, the smooth muscle myoblasts rapidly differentiate
into immature smooth muscle myocytes, but their final is essential for regional changes during morphogenesis
differentiation into mature myocytes does not occur un- and for cellular differentiation and patterning along the
til after birth.25,26 crypt-villus axis.9,32 This intricate process is the product
Role of the ENS in GI Development of tight spatial and temporal control of signaling mole-
cules and transcription factors in both germ layers.
It is beyond the scope of this review to discuss in For example, during mouse gastrulation, the early me-
detail the development of the ENS, which is organized soderm is patterned by the adjacent endoderm via se-
into the submucosal and myenteric plexuses. Each is creted signals. Fibroblast growth factor (FGF)-4 is one of
derived from neural crest cells, which migrate from the the earliest characterized signals33; its role persists after
neural plate between E9 and E13.28 –31 However, for the the formation of the gut tube, when it guides the adja-
purposes of this review, it is relevant to remember that as cent endoderm along the anterior-posterior axis, inhibit-
the gut develops, it is populated by migrating neural crest ing anterior fates and promoting posterior fates in a
cells that home to the mesenchymal layers. Genetic ma- concentration-dependent manner.34 The instructive role
nipulation of the developing gut often leads to a Hir- of FGF-4 is time sensitive, and as development proceeds
schsprung’s-like phenotype, suggesting that the integrity FGF-4 loses its ability to repress anterior development.34
of multiple pathways is required for normal innervation Shortly after gastrulation, possibly in response to en-
and muscle development of the gut. dodermal FGF signals, bone morphogenetic proteins
(BMPs) pattern the mesoderm of Xenopus embryos into
Development of the Lateral Plate its different components, including the VM. This exam-
Mesoderm ple of a dorsoventral morphogen in the mesoderm is
The molecular network that governs lateral plate illustrated in Figure 3.
mesoderm development depends on signals from the Interactions between Shh, BMP-4, and Foxf1 partici-
endoderm. Cross talk between mesoderm and endoderm pate in VM morphogenesis and appear to be conserved
2078 MCLIN ET AL GASTROENTEROLOGY Vol. 136, No. 7
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

Table 1. Gene Nomenclature


Drosophila Xenopus Mouse Chick Zebrafish Human
Biniou FoxF1 Foxf1/Foxf2 Not identified Not identified FOXF1
Dpp Smad Smad Smad Smad SMAD
Bagpipe Xbap; zampogna Nkx 3.2/Bapx1 Bapx1 bapx1 NKX3.2
Tinman Nkx2.3,2.5 Nkx2.3–2.7 Nkx2.5 nkx 2.3–2.7 NKX 2.3–2.9
vimar Not identified Not identified Not identified Not identified Not identified

NOTE. The names of homologous genes in the different species discussed in the text are summarized. The bagpipe family has 2 related but
different genes in Xenopus. In vertebrates, tinman has multiple homologues belonging to the Nkx2 cluster. Except for the transforming growth
factor ␤ pathway (Dpp/Smad), signaling pathways are not included because molecules referenced in the text have conserved names across
species. In the text, when referring to a gene in a general sense, the murine nomenclature is used. Bapx, bagpipe homeobox-containing gene
1 homologue; Fox, forkhead box.

across species (Figure 1). Studies in chicks have shown severe foregut, gallbladder, and lung malformations.38,43
that the endoderm Shh signal activates BMP-4 expression The closely related gene Foxf2 is differentially expressed
in the lateral plate mesoderm5 where Smads (intracellular during mouse GI development, with expression predom-
mediators of BMPs) regulate mesodermal proliferation inating in the posterior aspect of the developing gut.44,45
and later differentiation into smooth muscle.3,5,35 In Xe- This suggests some functional redundancy between Foxf1
nopus embryos, BMP-4 activates the expression of FoxF1, and Foxf2 in the distal mesenchyme, which would explain
a transcription factor required for lateral plate and VM the predominantly foregut phenotype of the FoxF1⫹/⫺
formation.36 Furthermore, it is sufficient to induce FoxF1 mice and distal phenotype of Foxf2⫺/⫺ mice. Consistent
expression in nonmesodermal tissue, and FoxF1 messen- with its expression pattern, Foxf2-null embryos have co-
ger RNA rescues the phenotype of embryos injected with lonic dilatation and anal atresia, whereas the foregut
a dominant negative BMP-4 receptor.36 In Drosophila, seems relatively unaffected.46 Although this phenotype is
Dpp (homologous to Smads in vertebrates; see Table 1) reminiscent of Hirschsprung’s disease in humans, no
signals are both upstream and downstream of biniou, the human condition has been associated with absence or
Foxf1 homologue essential for VM development.37 In mutations of FoxF proteins. In sum, FoxF proteins are
mouse, Foxf1 messenger RNA is absent in the foregut essential for early VM development and multiple aspects
structures of Shh⫺/⫺ embryos.38 Therefore, induction of of intestinal differentiation.
the VM via Foxf1 depends on endodermal Hh signals and To further characterize the molecular regulation of
mesodermal BMPs (see Table 1 for species-specific no- early VM development, Jakobsen et al used gene profiling
menclature). and ChIP-on-Chip (chromatin immunoprecipitation fol-
The Drosophila gene biniou and its vertebrate homo- lowed by microarray) to identify downstream targets and
logues Foxf1 and Foxf2 have emerged as master regulators partners of biniou in Drosophila.39 They showed that al-
of VM morphogenesis and differentiation across spe- though biniou is expressed throughout VM development,
cies.36,37,39 In biniou-null Drosophila embryos, VM deve- its binding to specific enhancer elements is under tight
lopment arrests before GI morphogenesis is complete37 temporal and spatial control. Importantly, they showed
(Figure 4A and B). Likewise, in Xenopus laevis, FoxF1 that several biniou targets are conserved in mice; the
knockdown leads to severe defects in gut coiling and expression of Tcf21, Sall4, and Ptk7 were down-regulated
elongation, partly because of decreased cell proliferation in Foxf1⫺/⫺ or Foxf2⫺/⫺ mouse embryos.39 Bagpipe and
in the mesoderm36 (Figure 4C and D). In mouse embryos, Mef2 are also important regulators of VM development
Foxf1 expression localizes to the interfaces between mes- and differentiation. Each can bind its own set of regula-
enchyme and epithelium40; knockout studies have shown tory elements in the absence of biniou.47 Understanding
a role in early lateral plate mesoderm formation and later the differences between biniou-dependent and -indepen-
differentiation. The earliest known function of Foxf1 is in dent genes will be an important avenue to explore to
specifying VM (splanchnic) from somatic mesoderm. In advance our understanding of regional and temporal cell
Foxf1-null embryos, there is incomplete separation be- fate decisions during VM development. Of course, it will
tween the visceral and somatic leaflets of the lateral plate; be crucial to confirm these findings in vertebrates.
the homeobox gene Iroquois is ectopically expressed in the Unlike some of the other transcription factors known
visceral leaflet, suggesting that Foxf1 specifies VM by to participate in regional differentiation, biniou/Foxf1 is
inhibiting Iroquois41 (Figure 1). Although Foxf1-null em- expressed throughout the length of the developing intes-
bryos die at midgestation because of defects in the vas- tine (Figure 5A and B). This is of interest because there is
cular development of the extraembryonic membranes,42 a wealth of genes known to participate in foregut and
abnormal esophageal development has occurred by this hindgut development, whereas there are few midgut can-
stage.41 Because both alleles of Foxf1 are required for didates. Further studies to address the differential re-
normal VM development, Foxf1⫹/⫺ embryos also have gional regulation of Foxf1 should elucidate whether the
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2079

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
development, across species, is that Hh ligands are
expressed by the endoderm/epithelium and that Hh re-
ceptors and targets are expressed by the mesoderm/mes-
enchyme, which responds differentially according to de-
velopmental stage and anterior-posterior position.4 – 8,48
Multiple signaling pathways confer positional cues to the
developing intestine (Figure 6). Because of their known
role in segmental and anterior-posterior patterning, ho-
meobox gene products are obvious candidates to inte-
grate these positional cues into regional signals in the

A
Nkx2.3-2.9

Nkx3.2

vimar FoxF1 zampogna

Wnt, FGF, RA

Foregut Midgut Hindgut


B
p
visceral
mesoderm
l liver p

Barx1
Nkx3.2
Hoxa-5
Hoxa-4 Hoxd-12/13

Foxf1/f2
Nkx2.3
Hlx
FoxL

Figure 5. Transcription factors and signaling pathways during anter-


ior-posterior and radial patterning. (A) Early mesoderm formation is
Figure 4. The role of biniou/FoxF1 is conserved across species. Inver- under the control of the conserved homeobox genes. Studies in Dro-
tebrate example: Drosophila melanogaster. (A) Expression of VM sophila have allowed the identification of some early steps in VM forma-
marker fascilin III (Fas III) in stage 12 Drosophila embryos (arrow). Ante- tion implicating homeobox genes.180,181 These include tinman (Nkx2.3-
rior is to the left. (B) The VM marker Fas III is absent in the biniou mutant, 2.9), bagpipe (Nkx3.2), and other genes.181 Tinman is required for
which lacks biniou, the Drosophila orthologue of FoxF1.37 Vertebrate mesoderm formation, whereas its downstream target bagpipe pro-
example: effect of FoxF1 loss of function in Xenopus laevis. (C) Ventral motes VM development.182,183 Vimar, a downstream factor from bag-
view of a normal gut coiling in a 5-day-old Xenopus embryo. (D) Both pipe, further specifies VM development.184 In Xenopus, 2 bagpipe ho-
coiling and elongation are severely impaired when FoxF1 is knocked mologues have been identified and their expression has been analyzed,
down using antisense morpholino-oligonucleotides.36 The black cells in suggesting that these the role of these homeobox genes is conserved in
C are melanocytes; they are absent in the knockdown embryos. Cranial development; Xbap (Nkx3.2) is expressed in the foregut musculature of
is to the top. Xenopus and mouse82,185 and zampogna in the musculature of the
Xenopus midgut, with some overlap in the posterior foregut meso-
derm.185,186 Expression of zampogna in the posterior indicates early
anterior-posterior differentiation in Xenopus. Vimar is indicated in green
molecular network that regulates midgut development is in the figure because it has been identified in Drosophila but not in
a “default” program that needs to be modified for the Xenopus. Foxf1, downstream of Nkx3.2 (bagpipe), is first expressed in
development of more recent digestive functions such as the late gastrula and then throughout the VM of the embryo and the
the stomach. What factors participate in regionalization adult. Although the exact relationship between these genes and major
signaling pathways is incompletely understood, in the early embryo,
of the VM?
FGFs, Wnts, and retinoic acid (RA) promote posterior fates while inhib-
iting anterior fates. Figure of Xenopus embryo at stage 28: © 1994
Anterior-Posterior Patterning of the Pieter D. Nieuwkoop and J. Faber.187 (B) Spatial distribution of selected
Mesoderm and GI Tract mesodermal forkhead and homeobox genes involved in vertebrate gas-
trointestinal differentiation, using mouse nomenclature. Not shown is
GI development progresses in a craniocaudal Nkx2.5, which specifies the pylorus in the caudal segment of the fo-
manner, but the mechanisms by which this occurs are regut. Anterior is to the left, dorsal to the top. Diagram courtesy of Aaron
not fully understood. The conserved paradigm of GI M. Zorn. l, liver; p, pancreas.
2080 MCLIN ET AL GASTROENTEROLOGY Vol. 136, No. 7
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

atresias58; Hoxa13 mutations have been described in hu-


mans with limb, GI, and genitourinary abnormalities.59,60
Misexpression of Hoxd13 in the chick midgut leads to a
hindgut phenotype (homeotic transformation). These
findings suggest that loss of Hox genes leads to malfor-
mation of the intestines of mice, whereas their ectopic
expression in chick embryos leads to a homeotic trans-
formation or ectopic expression of region-specific genes.
Together with the finding that Hoxa4⫺/⫺ mice have ab-
normal colonic musculature61 and Hoxa5⫺/⫺ mice have
abnormal stomach development,62 these findings indi-
cate that Hox gene products probably participate in re-
gionalization in part through the regulation of smooth
muscle precursor cell fate and proliferation.
Third, Hox genes likely confer positional information
along the mouth-to-anus axis by integrating multiple
Figure 6. Heterotopic recombinations of mesoderm and endoderm. regulatory inputs. In the early mouse embryo, BMP sig-
Detailed dissection and recombination experiments in neurula-stage naling in the VM is necessary for Hox gene induction;
embryos (during regional specification of the primitive gut tube) of Xe-
nopus laevis, the African clawed frog (equivalent to mice with 7– 8
subsequent mesenchymal-to-epithelial signaling controls
somites or a 22-day-old human embryo), have shown that early stomach morphogenesis and lung budding,63 and reti-
endodermal regionalization depends on mesodermal signals.188,189 Al- noic acid signaling controls endodermal expression of
though the nature of these signals is incompletely understood, the Hox genes such as Pdx1 (pancreatic and duodenal ho-
major signaling pathways appear to be involved. For example, in the
meobox 1) or Cdx2 (caudal type homeobox transcription
chick, the endoderm needs instructive BMP/activin family signals from
the lateral plate mesoderm to differentiate64; retinoic acid (RA) in the VM factor 2).64 Cdx genes are regulated by Wnts, and it is
confers a posteriorizing gradient to the developing GI tract.190 In mouse, hypothesized that the Wnt pathway “co-opted” Hox ma-
Xenopus, and zebrafish, mesodermal Wnt signals confer temporally chinery to execute their anterior-posterior program.65– 69
regulated anteroposterior information to the adjacent endoderm.191,192 Further studies are required to determine if this holds
Wnt antagonists in the anterior VM are necessary for stomach devel-
opment (anterior)83 and Wnts probably posteriorize the VM, but this has
true in VM development. Finally, considering that the
not been shown. Because this paradigm has been studied in detail in mesoderm appears to respond differentially to Hh signals
liver and pancreas development, we refer the reader directly to those along its anterior-posterior axis, Hh might induce me-
studies.191–193 Modified and reprinted with permission from Horb and sodermal Hox gene products to induce sphincter for-
Slack.189
mation. This is illustrated by Shh⫺/⫺ mice that display
homeotic transformation phenotype: intestinal transfor-
mation of the stomach, annular pancreas, duodenal ste-
presumptive intestine (Figure 5A). Detailed expression
nosis, and an imperforate anus. This homeotic transfor-
studies have mapped Hox gene products in the develop-
mation of the Shh⫺/⫺ stomach supports the concept of an
ing gut; the majority are expressed in the mesoderm.49 –53
Hh-to-Hox pathway, which has now been demonstrated
There are 3 important points about the role of Hox
gene products in GI patterning. The first is that their role in the foregut and hindgut of chick embryos, mice, and
appears to have been selected for during evolution. Ho- rats.5,62,70
meodomain-containing genes belonging to the Nkx2 Although Shh becomes restricted to the foregut and
family (Figure 5A) confer positional information.54 Over- hindgut after E14.5, Ihh is expressed throughout the
expression of vertebrate Nkx2 genes can partially rescue length of the intestinal epithelium during development
the phenotype of Drosophila embryos null for the tinman and adulthood.6 These expression patterns are consistent
gene in the same cascade, indicating a highly conserved with the phenotypes of knockout animals.6,7 In contrast
function for these genes (tinman is upstream of Nkx2; to the Shh⫺/⫺ phenotype, Ihh⫺/⫺ mice have a phenotype
Figure 5A).55 that resembles human Hirschsprung’s disease, nearly ab-
Another take-home point concerning Hox genes is that sent enteric neurons and a thin muscularis propria,71
they confer regional information that contributes to the which is also observed in the Foxf2⫺/⫺ mice.46 Although
formation of boundaries by conveying permissive signals an Hh-to-Fox axis has not been explored in detail, the
to the mesenchyme.3 In mice carrying a homozygous role of Fox genes in this process has been supported by
deletion of the Hoxd3-13 cluster,56 which is normally lack of Foxf1 expression in the GI tract of mice with Gli2
expressed in the mesoderm, sphincter formation of both and Gli3 mutations.72 Importantly, these phenotypes
the pylorus and anus is absent.56 Furthermore, ectopic mimic human syndromes with GI malformations that
expression of Hoxd12 and Hoxd13 causes abnormal or have been associated with Hh pathway defects, such as
absent ceca.57 Hoxa13⫺/⫺ mice develop GI stenoses and Pallister–Hall syndrome73 and the VATER association.74
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2081

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
Mesenchyme-to-Endoderm Signaling in Therefore, the hindgut endoderm, like its anterior coun-
Stomach Development terpart, shows relative plasticity late in development in
Stomach development is a unique example of the response to instructive signals from the VM. ISEMFs
differential response of the mesenchyme-to-endodermal appear to have a role in this process; ISEMFs from an-
signals. The stomach of the developing chick has an teroposterior segments of the intestine secrete varying
anterior and a posterior chamber. Recombination exper- amounts of growth factors, thereby conferring dose-
iments of proventriculus (anterior) mesenchyme and giz- dependent regional characteristics to the adjacent
zard (posterior) epithelium (and vice versa) have shown endoderm.19 Similar experiments in chicks showed that
that the epithelium often adopts the fate of the adjacent recombination of proventricular mesenchyme with mid-
mesenchyme.3,75,76 However, this is not the case in other gut endoderm did not alter epithelial gene expression in
parts of the GI tract.77–79 Signaling by BMPs 2 and 4 the midgut but did in the foregut.90 Taken together,
participate in this cross talk, although their relative con- these findings suggest that the midgut endoderm pro-
tribution differs according to developmental stage.80,81 gram might be specified earlier than the cranial- and
The divergent homeobox-containing gene Bapx1 or caudal-most segments of the gut.
Nkx3.2 regulates mesodermal BMP-4 and Wnt5a expres-
sion in the chick stomach75 and pylorus morphogenesis The Lateral Plate Mesoderm and
in the mouse82 (Figure 6). In mice, the regionally re- Mesenchyme in Left-Right Asymmetry
stricted homeodomain-containing protein Barx1 is re- of the GI Tract
quired for mesenchymal expression of Wnt inhibitors In all vertebrates, shortly after gastrulation, the
and normal development of the stomach epithelium.83 In lateral plate mesoderm participates in left-right cell fate
chick, mesodermal BMP-4 independently induces expres- decisions. The transforming growth factor ␤ family
sion of Nkx2.5 and Sox 9 in a cell-autonomous manner, member Growth and differentiation factor-1 (Gdf-1)91 pro-
leading to the formation of the pyloric sphincter.84,85 motes the expression of genes that regulate lateral devel-
Consistent with the role of BMP in anterior-posterior opment, such as nodal, Lefty, and Pitx2 in the left lateral
patterning, constitutive activation of BMP signaling in plate mesoderm.92–96 Recently, events downstream of
distal segments of the developing gut mesenchyme leads Pitx2 (paired-like homeodomain transcription factor 2)
to ectopic, cell-autonomous expression of the pyloric and its partner LIM-homeodomain containing transcrip-
sphincter marker Nkx2.5.86 In summary, regulation of tion factor Isl1 (islet-1) were identified in the dorsal
major growth factors by mesodermal homeobox genes is mesentery. The dorsal mesentery, which is continuous
necessary for epithelial stomach morphogenesis and for- with the visceral mesenchyme, derives from the lateral
mation of the pylorus. These observations raise 2 impor- plate mesoderm and anchors the gut to the dorsal wall of
tant points. First, regionalization of the developing GI the vertebrate embryo. Pitx2 and Isl1 control expression
tract is governed in part by circumscribed transcription of glycosaminoglycans and the ECM protein N-cadherin;
factors in response to local signals.83 Second, precise expression is asymmetrically distributed in the dorsal
regulation of signaling pathways in the stomach suggests mesentery of chick and mouse, leading to changes in cell
that gastric development requires modification of the shape and intercellular connections that induce the first
midgut “default” program. From a developmental per- gut tilt to the left.97 This study has shown a role for the
spective, it seems both economical and practical to mesoderm-derived mesentery in gut morphogenesis. Be-
modify a preexisting program for the development of a cause Shh⫺/⫺ and Ihh⫺/⫺ mice display malrotation, it is
new structure with slightly different functional re- probable that endodermal Hh signals are upstream of
quirements. this pathway.71 In Xenopus, retinoic acid–metabolizing
enzymes are expressed in the VM of the gut during the
Midgut and Hindgut Development early stages of gut coiling.98 Addition of retinoic acid or
Unlike the stomach, the plasticity of the midgut retinoic acid inhibitors to embryonic culture medium
epithelium, in response to changing mesenchymal sig- before the onset of gut coiling leads to abnormal chirality
nals, appears to be somewhat limited.87,88 Using hetero- and organ heterotaxy.98 Thus, the Gdf-1/Pitx2/ECM
topic cross-associations of endoderm and mesenchyme pathway likely results from the integration of multiple
from different segments of the developing rat intestine, signaling gradients in the mesenchyme.
Ratineau et al showed that segments of intestinal mes-
enchyme from rat fetuses did not confer equal regional The Role of the VM in Radial
information to adjacent endoderm. In the 14-day-old Patterning, Villus Formation, and the
fetus, midgut endoderm did not change its enzyme ex- Differentiation of the Epithelial and
pression when cocultured with proximal or distal intes- Mesenchymal Layers
tinal mesenchyme.89 Conversely, colon endoderm did In addition to its role in anterior-posterior and
respond to the association with jejunal or ileal mesen- left-right patterning, the mesoderm is required for radial
chyme by developing into small intestinal epithelium. patterning and villus formation. Radial patterning is the
2082 MCLIN ET AL GASTROENTEROLOGY Vol. 136, No. 7
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

Table 2. Transcription Factors in Radial Patterning of the Gut


Transcription factor Model Expression Phenotype References
FoxF1 Xenopus VM Absent lumen, absent smooth muscle actin expression 36
Foxf1⫹/⫺ Mouse VM, foregut Esophageal atresia, tracheo-esophageal fistula 38, 41
Foxf2⫺/⫺ Mouse VM Abnormal muscularis propria, ECM, absent ENS, 38, 41
dilated colon, increased epithelial proliferation
Hlx⫺/⫺ Mouse VM Epithelial proliferation, ENS abnormalities, short gut 195–197
Nkx2.3⫺/⫺ Mouse, chick VM midgut and hindgut Thin mesenchyme, abnormal villus formation 198, 199
Hoxa5⫺/⫺ Mouse VM midgut ENS, smooth muscle, ECM 62

organization of the GI tract into concentric layers.80 Signaling Pathways in Radial Patterning
Differentiation of the crypt-villus axis happens at differ- Multiple cell fate and positional decisions contrib-
ent times in development in different species and ute to the formation of the organized, tubular gut. To
progresses in a craniocaudal fashion. In mice, villus for-
this end, transcription factors regulate and integrate nu-
mation begins around E15, but crypt formation does not
merous secreted signals, leading to the differentiation of
occur until the postnatal period. In Xenopus, the trough/
each mesodermal component and its orientation along
intestinal fold axis is the product of metamorphosis, and
the crypt-villus or epithelium-serosa axes. Although these
in humans, the newborn infant has a complete crypt-
villus axis. Crypt-villus axis formation relies largely on different signaling pathways overlap and their relative
mesenchymal-epithelial interactions that determine vil- contribution is still incompletely understood, we will
lus length, crypt depth, and spacing between villi.77,99 –101 now discuss their individual roles as they are character-
The inaugural event in villus formation is mesenchymal ized to date.
condensation, which depends on ubiquitous signaling Hh: smooth muscle proliferation and positioning
pathways that vary according to the developmental time of the crypt. Loss-of-function studies of different com-
period. Certain transcription factors that participate in ponents of the Hh pathway have led to the comparatively
early morphogenetic processes also contribute to radial well-understood role of this pathway in the radial pat-
patterning and villus formation. terning of the GI tract. In very broad terms, its functions
are 4-fold, keeping in mind, however, that these vary
Transcription Factors That Contribute to according to developmental window. First, Hh ligands
Radial Patterning act as radial morphogens for the development of the
The transcription factors Foxf1/f2 are expressed smooth muscle layer; in other words, too much or too
throughout development; apart from their roles in early little ligand inhibits smooth muscle development, while
patterning of the lateral plate, they also have a role in the right concentration, regulated by the distance from
later differentiation.36,46 Studies of mouse knockout the endodermal source, induces smooth muscle forma-
models have shown that Foxf proteins are required for tion in both chick and mouse.35,71 Second, depending on
multiple aspects of VM development, including radial developmental window, endodermal Hh signals are im-
patterning. Foxf1/2 expression in the early VM is ubiq- portant for induction of reciprocal signaling from the
uitous and Foxf proteins are required for VM develop- mesenchyme back to the endoderm, because loss of func-
ment in multiple species. However, because of the pleo-
tion leads to epithelial proliferation.102 Third, epithelial
morphic phenotypes of knockout animals, it is difficult
Hh ligands influence ISEMF localization along the crypt-
to determine the precise role of Foxf genes. Several other
villus axis, probably because these cells serve to integrate
transcription factors participate in radial patterning,
the epithelial signal, thereby regulating the proliferative
with their respective roles still incompletely understood.
effect of the Hh signal.103 Fourth, Shh and Ihh have
The difficulty in understanding the precise role of a given
transcription factor stems from the developmental inter- complementary roles in ENS development; Shh inhibits
dependence of mesenchymal components among them- ENS development, while Ihh is essential.35,71 In summary,
selves and with the adjacent epithelium. Table 2 summa- the Hh pathway serves as a radial morphogen affecting
rizes the phenotypes associated with loss of function of both cell fate and proliferative decisions in the layers of
forkhead or homeobox-containing transcription factors the mesenchyme and epithelium. In the future, targeted,
in the VM. Smooth muscle, enteric neurons, and ECM tissue-specific modification of Hh signaling to the mes-
appear to be the most vulnerable components of the enchyme should reveal the relative contributions of Ihh or
developing VM in response to genetic manipulation. In Shh to the development of the different concentric layers.
addition to changes in the VM, genetic manipulation of Further, it may help elucidate the concentration and
the VM usually leads to aberrant epithelial development temporal needs of each cell type, thereby facilitating the
and proliferation. direction of cell fate in vitro.
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2083

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
BMPs: smooth muscle development and regula- BMPs, previously discussed for their role in anterior-
tion of the proliferative compartment (crypts). BMPs are posterior patterning, are also necessary for radial pattern-
members of the transforming growth factor ␤ superfam- ing and villus formation. In simple terms, BMP-4 in the
ily; BMPs 2 and 4 have important roles during GI devel- VM controls smooth muscle proliferation and differen-
opment: signaling via the BMP receptor (BMPR)-1a.80,104 tiation throughout most of the developing vertebrate
BMP transcription in the VM is initiated in response to GI tract107; this is probably conserved across species.
endodermal Hh signals,5 so many of the BMP-related Whereas the small intestine needs sufficient expression of
phenotypes are reminiscent of Hh loss-of-function phe- BMP-4 in the mesenchyme for the development of an
notypes discussed in the previous section. Although the adequate muscularis propria, overexpression of BMP-4 in
role of BMPs in GI development also varies according to the mesenchyme of the embryonic chick gut delays dif-
developmental stage and tissue layer, studies in the early ferentiation into smooth muscle106 and inhibition of
developing gut of avian embryos suggest that BMP sig- BMP-4 in the stomach is required for the highly devel-
nals are required by all gut layers that are distal to the oped musculature to meet its functional needs.106 Ab-
stomach105 and contribute to the morphogenesis and sence of BMP-4 from the presumptive hindgut meso-
differentiation of all components of the GI tract.106 derm promotes aberrant endoderm proliferation and
ENS development, indicating the importance of BMP-4
not only in smooth muscle development but also in the
regulation of epithelial proliferation.81
BMP signaling is important for villus formation and
endodermal/epithelial patterning (Figure 7A and B). Its
role in epithelial morphogenesis and differentiation is
complex and involves cross talk between epithelium and
mesenchyme; several studies have attempted to address
this issue.106 –109 When the secreted BMP inhibitor nog-
gin is constitutively expressed in the developing mouse
epithelium, an excess number of crypts develop.107,110
Depending on the model and the developmental stage
analyzed, epithelial and mesenchymal proliferation and
differentiation are affected, leading to aberrant crypt and
villus formation, similar to that associated with the
pathogenesis of human juvenile polyposis.110 However,
when BMP signals to only the epithelium are specifically
blocked using a villin-dnBMPR1a (dominant negative BMP
receptor 1a) transgene, only epithelial proliferation and
secretory lineage differentiation are affected; villus or
crypt number are not.109 Taken together, these studies
support a model in which BMP signaling in the mesen-
chyme sends antiproliferative signals to the crypts and
epithelial stem cells, probably through a signaling relay
in the pericrypt mesenchyme (possibly ISEMFs), that
regulates the size and location of the proliferative com-
partment.108 It is unclear whether the mesodermal BMP
brake is conserved across species and whether it is in
effect at all developmental time points. In contrast to the
evidence presented from mice, during gut remodeling
associated with Xenopus metamorphosis, mesenchymal
BMPs promote rather than inhibit epithelial prolifera-
Figure 7. Molecular cross talk between the VM and the endoderm tion.111,112 The obvious next step in elucidating the re-
during crypt-villus formation and in the adult. (A) Signaling pathways
during crypt-villus formation. Hh ligands signal to the mesenchyme. In
lative roles of BMP signaling in the epithelium and
response, probably via ISEMFs, mesodermal Wnt ligands promote pro- mesenchyme is the conditional modification of BMP
liferation in the intervillus space and in the villus (starting at E16.25) and signaling in the mesodermal layer, something impeded
BMP signals regulate proliferation of the epithelium. Arrows indicate by lack of appropriate tools.
direction of signal. (B) In the adult with a fully developed crypt-villus axis. Wnts: proliferation versus differentiation. Wnts
On the left, pathways expressed in the mesenchyme; on the right,
pathways expressed in the epithelium. The signals in the adult likely
are accepted as the master regulators of epithelial prolif-
occur simultaneously. Modified and reprinted with permission from eration and differentiation in the developing and adult
Crosnier et al.194 GI tract.113–115 Most studies have focused on the role of
2084 MCLIN ET AL GASTROENTEROLOGY Vol. 136, No. 7
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

Wnts in endoderm specification and differentiation, be- with aberrant epithelial expression of nuclear ␤-catenin
cause regulation of nuclear ␤-catenin accumulation is and increased epithelial proliferation. Histologically,
required for epithelial homeostasis and carcinogene- Foxl1-null mice have delayed formation of intestinal villi
sis.83,116,117 The origin of these Wnt signals is mostly compared with wild-type mice, but this is not observed in
mesodermal118,119 (Figure 7A and B). The exact cellular Foxf mutant mice, in which the mesodermal structures
origin of the Wnt signal in vivo is still unclear, but appear to be normal.127,129 Adult Foxf mutant mice have
ISEMFs are likely to contribute because they express Wnt multiple epithelial abnormalities, including increased cell
ligands and because they are adjacent to the proliferative proliferation in the villi, cystic inclusions, and abnormal
compartment of the epithelium, the intestinal crypts.118 stomach histology.127,129 Loss of Foxl1 augments the ep-
In situ hybridization analyses of mouse and chick ithelial effect of adenomatous polyposis coli (APC) germline
embryos have shown that the mesenchyme of the devel- mutations by increasing the multiplicity of intestinal
oping and adult intestine contains high levels of Wnt adenomas.128 The developing intestine expresses Wnt li-
pathway components, both canonical and noncanoni- gands, receptors, and inhibitors, and their roles vary
cal.119,120 In the adult, Wnt signals are necessary for during development. The control of mesodermal Wnt
maintenance of the proliferative compartment of the signals is of particular importance because of the contri-
epithelium.121 During development, it has been accepted bution of these factors to epithelial tumorigenesis. Tis-
that the intervillus mesenchyme expresses Wnt compo- sue-specific modulation of Wnts in the mesenchyme will
nents because signs of Wnt activity were found in the improve our understanding of the role of Wnts both in
epithelium.122,123 However, novel findings suggest the the mesenchyme proper and in the adjacent epithelium.
adult pattern of crypt-predominant Wnt activity only FGFs. The role of the FGF pathway in radial
begins around postnatal day 3 in mice; before this time differentiation is less well understood that that of the
point, canonical Wnt activity predominates in the nas- Hh, BMP, or WNT signaling pathways. In Xenopus, FGF
cent villi coincident with the mesenchymal expression of signaling is necessary for expression of the smooth mus-
several Wnt ligands, including Wnt5a119,124 (Figure 7A). cle cell marker XSM22␣130 and smooth muscle actin and
Importantly, in this study, ␤-catenin activity does not patterning of the early intestine.131 In the E18.5 mouse
correlate with epithelial proliferation, suggesting that embryo, FGF13 is highly enriched (47.6-fold) in the mes-
Wnt signaling may be more than just an on/off switch for enchyme compared with epithelium132 and likely medi-
proliferation versus differentiation.124 ates autocrine actions via FGFR1 and R2, as well as
Recent studies have highlighted multiple roles for having a role in crypt morphogenesis via FGFR3133 (Fig-
Wnt5a in the mesenchyme.119,125 Wnt5a is expressed both ure 7B). Normal cecum development is characterized by
in the embryonic and adult mesenchyme, suggesting that an outward growth of both mesenchymal and epithelial
it is an important signaling molecule in intestinal devel- proliferation134 and requires mesenchymal FGF-10 and
opment and homeostasis, probably with different spatial BMP-4 signaling in response to epithelial FGF-9 sig-
and temporal functions, acting through both the canon- nals.134 Mouse studies indicate that epithelial FGF-9 sig-
ical and noncanonical pathways.119 Wnt5a⫺/⫺ mice have naling controls gut length by regulating proliferation
multiple defects: thinner muscularis propria, improper and differentiation of subepithelial fibroblasts, possibly
midgut closure, and a dramatically shortened midgut. through putative mesenchymal stem cells.126 FGFs are
This latter effect is in part mediated by defective post– required for normal development of the gut because they
mitotic cell intercalation in the epithelium in a non—␤- regulate mesenchymal signaling, but we need to learn
catenin– dependent manner.125 This finding joins that of more about the time periods and regions in which these
others in highlighting a role for the mesenchyme in gut potent signaling molecules are active or inactive. Like
elongation and morphogenesis125,126 and highlights the other pathways, the understanding of their relative con-
importance of Wnt signaling in more than epithelial tribution to mesodermal and epithelial patterning will be
proliferation and differentiation. vastly aided by the development of mesoderm-specific
Together with other Wnt ligands, Wnt5a is also essen- tools.
tial during radial patterning when it cooperates with Hh,
BMPs, and Forkhead genes. As outlined previously, Foxf Glucocorticoid and Thyroid Hormones
mutants have altered collagen production and decreased in GI Development
mesenchymal BMP4 signaling, which leads to Wnt5a Glucocorticoids (GCs) promote maturation and
overexpression and increased epithelial proliferation.46 differentiation of the human and rodent intestinal epi-
Similarly, Foxl1 participates in the regulation of Wnt thelium.87,135 However, the mesenchyme appears to be
signals to the adjacent epithelium. Although Foxl1 posi- essential for this effect. In vitro studies have shown that
tively regulates BMP-4 and BMP-2 expression, its main fetal rat intestinal endoderm cells do not respond to GCs,
mechanism of action involves proteoglycan synthesis in but when they are cultured with mesenchymal cells and
the mesenchyme.127,128 Ectopic syndecan-1 synthesis in GCs, ␣-glucosidase expression is induced.136,137 Further-
the villus mesenchyme of Foxl1-null mice is associated more, although explants of intact perinatal colon do not
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2085

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
respond to GCs, sucrase expression is induced when Cytokines and Other Signaling
explants derived from colonic endoderm are associated Molecules in GI Development
with small intestinal mesenchyme and exposed to GCs.138 In addition to modulators of the major signaling
These findings indicate that small intestinal mesen- pathways and hormones, the VM of E18.5 mouse em-
chyme, but not colonic mesenchyme, enables GC induc- bryos is enriched in multiple peptide growth factors and
tion of maturation in an associated epithelium. Mesen- cytokines.132 These include endothelins, insulin-like
chymal cells alone respond to GCs; these cells express the growth factor (IGF), and platelet-derived growth factor
GC receptor137 and show increased expression of collagen (PDGF). For example, PDGF is believed to regulate mat-
type IV messenger RNA139 following in vivo administra- uration of smooth muscle precursors via the Wnt path-
tion of GCs.139 –141 In addition, in vitro studies have way.152 PDGF-A and its receptor PDGFR␣ contribute to
shown that in response to GCs, mesenchymal cells de- formation of the crypt-villus axis.153 They are both ex-
posit laminin at the mesenchymal-epithelial interface, pressed in the developing mouse small intestine starting
which appears to be essential for the effects of GCs on around E15.5; PDGF-A is expressed in the epithelium,
the epithelium because the effects can be blocked in vitro and PDGFR is expressed in the mesenchyme. PDGF-A⫺/⫺
by the addition of anti-laminin antibodies.142 These ex- and PDGFR␣⫺/⫺ mice have similar phenotypes; com-
periments suggest that the mesenchyme acts as either a pared with wild-type mice, they have fewer and broader
direct GC target that signals to the epithelium or that the villi, a thinner mesenchyme, and premature expression of
mesenchyme induces the adjacent epithelium to respond smooth muscle actin in the villus mesenchyme.153 There-
to the hormone.142 Regardless of the specific mechanism, fore, PDGF-A must promote cell proliferation and pre-
questions of clinical significance are whether the mesen- vent premature differentiation of the mesenchyme.
chyme is responsible for the relatively narrow develop- Again, the role of this molecule depends on the develop-
mental stage during which the intestinal epithelium can mental stage, because mutations in the PDGFR␣ are
respond to GCs143,144 and whether these mesenchymal associated with GI stromal tumors in humans.154 –158
effects are permissive or instructive. IGF-1 is expressed in the mesenchyme of the E18.5
Thyroid hormone also has a role in the development of intestines of mice,132 and IGF-1 receptors are expressed in
the vertebrate GI mesenchyme. Thyroid-responsive genes the submucosal region of the neonatal small intestine;
are expressed in the mesenchyme of E18.5 mouse embr- expression is down-regulated after birth.132,159 IGF-1 and
yos,132 and absence of the thyroid hormone receptor T3R␣ IGF-2 binding to the IGF receptor promotes epithe-
leads to hypoplastic smooth muscle layers in the devel- lial differentiation.159 Suppressor-of-cytokine signaling
oping intestine.145 This finding correlates with increa- (SOCS) 2 is normally required for regulating the effect of
sed expression of T3R␣ in the intestinal muscle layers IGFs. Consistent with this role, SOCS2-null mice have
of wild-type animals,145,146 which might suggest that increased size and weight, intestine length, and thickness
smooth muscle cells require a thyroid signal for develop- of the lamina propria.160 Members of the SOCS family
ment. The role for thyroid hormone in mammalian epi- are highly expressed in the E18.5 mesenchyme,132 sug-
thelial maturation has been difficult to assess because gesting that, like other growth factors, IGF and cytokine
changes in thyroid status cause concomitant changes in signaling in the mesenchyme is tightly regulated.
circulating GCs.87 However, thyroid hormone can syner- The peptide endothelin-1 and its receptors are also
gize with GCs in eliciting developmental changes in the expressed in the E18.5 mesenchyme of mice.132 The co-
epithelium,147,148 but it is not known whether the mes- lonic mesenchyme of rat embryos expresses high levels of
enchyme has a part in this synergy. Xenopus species are endothelin 3 at E16.5, which promotes differentiation of
characterized by a thyroid hormone– driven metamor- adjacent epithelia in a dose- and region-dependent man-
phosis during which the GI tract undergoes remodeling ner.161
characterized by extensive changes of the mesenchyme Epimorphin (encoded by Stx2) is also up-regulated in
and development of the muscular layers. Although this the mesenchyme,132,162,163 where it is secreted by ISEMFs
developmental step is unique to anurans, it is an attrac- to mediate epithelial morphogenesis and control intesti-
tive model to examine intestinal mesoderm/mesenchyme nal length.162,163 Stx2-null adult mice have longer intes-
development; in 7 days, the 2-cell-thick VM proliferates tines and colons than their wild-type counterparts.162
into longitudinal and circular muscle layers as well as Three-week-old Stx2-null pups have increased crypt cell
numerous other cell types.146 Cultures of Xenopus intes- number and increased villus length, suggesting that epi-
tinal epithelial cells are insensitive to thyroid hormone– morphin controls cell proliferation during the final
induced apoptosis when they are cocultured with ECM stages of crypt-villus axis formation. This effect has been
molecules,149,150 suggesting that the ECM confers sur- proposed to result from decreased BMP-4 signaling and
vival signals to the epithelium. Therefore, the study of GI increased expression of ␤-catenin target genes.162 It is
remodeling during Xenopus metamorphosis should offer unclear why loss of the protein confers a regenerative
important insight into the development of the VM and advantage, but therapeutic manipulation of this pathway
its components.151 may be attractive to pursue.
2086 MCLIN ET AL GASTROENTEROLOGY Vol. 136, No. 7
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

Smooth muscle genes are up-regulated in the mesen- developing cell-based therapies for treatment of these
chymal compartment,132 and studies in zebrafish have disorders.
shown that visceral smooth muscle cells are involved in
GI ontogeny.27,164 Zebrafish expressing a mutant smooth References
muscle myosin heavy chain have abnormal smooth mus-
1. Gilbert SF. Developmental biology. Sunderland, MA: Sinauer
cle development, defects in formation of the posterior- Associates, Inc, 2000.
most segment of the intestine, uncontrolled epithelial 2. Yasugi S. Role of epithelial-mesenchymal interactions in differ-
proliferation, and aberrant epithelial expression of ECM entiation of epithelium of vertebrate digestive organs. Dev
proteins.164 This phenotype is similar to that of Ihh⫺/⫺ Growth Differ 1993;35:1–9.
and Foxf2⫺/⫺ mice. It is possible that smooth muscle 3. Roberts DJ, Smith DM, Goff DJ, et al. Epithelial-mesenchymal
myosin heavy chain is a downstream target of a putative signaling during the regionalization of the chick gut. Develop-
ment 1998;125:2791–2801.
Ihh-Foxf pathway that controls GI radial differentiation. 4. Ekker SC, McGrew LL, Lai CJ, et al. Distinct expression and
Matrix metalloproteinase molecules are expressed in shared activities of members of the hedgehog gene family of
the mesenchyme of E18.5 mouse embryos,132 and high Xenopus laevis. Development 1995;121:2337–2347.
levels of matrix metalloproteinases are transcribed in the 5. Roberts DJ, Johnson RL, Burke AC, et al. Sonic hedgehog is an
mesenchyme during intestinal remodeling in Xenopus em- endodermal signal inducing Bmp-4 and Hox genes during induc-
bryos (during metamorphosis). Matrix metalloprotein- tion and regionalization of the chick hindgut. Development
1995;121:3163–3174.
ases cleave various ECM components and are target genes 6. Bitgood MJ, McMahon AP. Hedgehog and Bmp genes are coex-
for thyroid hormone; therefore, they are likely to regulate pressed at many diverse sites of cell-cell interaction in the
ECM composition, basement membrane fenestrations, mouse embryo. Dev Biol 1995;172:126 –138.
and subsequent signaling to the adjacent epithelium dur- 7. Echelard Y, Epstein DJ, St-Jacques B, et al. Sonic hedgehog, a
ing remodeling.165,166 Although the role of matrix metal- member of a family of putative signaling molecules, is impli-
loproteinase in mammalian GI development is poorly cated in the regulation of CNS polarity. Cell 1993;75:1417–
1430.
understood, amphibian models offer insight into poten-
8. Stolow MA, Shi YB. Xenopus sonic hedgehog as a potential
tial mechanisms. morphogen during embryogenesis and thyroid hormone-depen-
dent metamorphosis. Nucleic Acids Res 1995;23:2555–2562.
Conclusions 9. Kedinger M, Simon-Assmann P, Lacroix B. [Development of the
digestive function: regulation of the maturation of intestinal
The VM is a complex tissue that gives rise to the
brush border enzymes]. Reprod Nutr Dev 1986;26:691–702.
smooth muscle, mesenchyme, and other cells that com- 10. Apelqvist A, Ahlgren U, Edlund H. Sonic hedgehog directs spe-
pose the outer layers of the mature intestine and has cialised mesoderm differentiation in the intestine and pancreas.
multiple essential functions during the development of Curr Biol 1997;7:801– 804.
the GI tract. The development of each VM cell type is 11. Winnier G, Blessing M, Labosky PA, et al. Bone morphogenetic
intricately linked to that of its neighbors, and although protein-4 is required for mesoderm formation and patterning in
the mouse. Genes Dev 1995;9:2105–2116.
many of the molecular interactions are incompletely
12. Wells JM, Melton DA. Vertebrate endoderm development. Annu
characterized, abnormal mesenchymal signaling almost Rev Cell Dev Biol 1999;15:393– 410.
invariably leads to aberrant epithelial proliferation. The 13. Zaret KS. Regulatory phases of early liver development: para-
VM and its components confer instructive and permis- digms of organogenesis. Nat Rev Genet 2002;3:499 –512.
sive signals to the adjacent developing epithelium; this 14. McLin VA, Zorn AM. Molecular control of liver development. Clin
instructional dominance appears to be mediated by sol- Liver Dis 2006;10:1–25, v.
15. Cano DA, Hebrok M, Zenker M. Pancreatic development and
uble factors and signaling pathways that regulate devel-
disease. Gastroenterology 2007;132:745–762.
opment of the mesoderm and endoderm. 16. Montgomery RK, Mulberg AE, Grand RJ. Development of the
Compared with the foregut and hindgut, the develop- human gastrointestinal tract: twenty years of progress. Gastro-
ment of the midgut is less well understood; it responds to enterology 1999;116:702–731.
different mesodermal signals. The developmental plastic- 17. Joyce NC, Haire MF, Palade GE. Morphologic and biochemical
ity of different GI segments is controversial, which could evidence for a contractile cell network within the rat intestinal
mucosa. Gastroenterology 1987;92:68 – 81.
be the result of the different models and conditions used
18. Sappino AP, Dietrich PY, Skalli O, et al. Colonic pericryptal
in experiments. Alterations in mesodermal gene expres- fibroblasts. Differentiation pattern in embryogenesis and phe-
sion patterns lead to abnormal mesoderm differentiation notypic modulation in epithelial proliferative lesions. Virchows
but also to epithelial proliferation, so one of the major Arch A Pathol Anat Histopathol 1989;415:551–557.
roles of the VM appears to be control of epithelial pro- 19. Plateroti M, Rubin DC, Duluc I, et al. Subepithelial fibroblast cell
liferation. Studying the molecular basis of GI develop- lines from different levels of gut axis display regional character-
mental plasticity and the control of epithelial prolifera- istics. Am J Physiol 1998;274:G945–G954.
20. Hall PA, Coates PJ, Ansari B, et al. Regulation of cell number in
tion by the mesenchyme could identify new therapeutic the mammalian gastrointestinal tract: the importance of apo-
targets for cancer and other diseases of the GI tract. ptosis. J Cell Sci 1994;107:3569 –3577.
Improving our knowledge of the spatial and temporal 21. Wallace KN, Pack M. Unique and conserved aspects of gut
control of GI cell fates might ultimately contribute to development in zebrafish. Dev Biol 2003;255:12–29.
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2087

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
22. Smith VV, Milla PJ. Histological phenotypes of enteric smooth gall bladder development. J Biol Chem 2002;277:12369 –
muscle disease causing functional intestinal obstruction in 12374.
childhood. Histopathology 1997;31:112–122. 44. Aitola M, Carlsson P, Mahlapuu M, et al. Forkhead transcription
23. Hohmann B, Holzwarth M, Saur G, et al. Neonatal necrotizing factor FoxF2 is expressed in mesodermal tissues involved in
enterocolitis and co-existing defect of the intestinal muscula- epithelio-mesenchymal interactions. Dev Dyn 2000;218:136 –
ture. Eur J Pediatr 1993;152:540 –541. 149.
24. Izraeli S, Freud E, Mor C, et al. Neonatal intestinal perforation 45. Ormestad M, Astorga J, Carlsson P. Differences in the embry-
due to congenital defects in the intestinal muscularis. Eur J Pe- onic expression patterns of mouse Foxf1 and -2 match their
diatr 1992;151:300 –303. distinct mutant phenotypes. Dev Dyn 2004;229:328 –333.
25. McHugh KM. Molecular analysis of smooth muscle development 46. Ormestad M, Astorga J, Landgren H, et al. Foxf1 and Foxf2
in the mouse. Dev Dyn 1995;204:278 –290. control murine gut development by limiting mesenchymal Wnt
26. McHugh KM. Molecular analysis of gastrointestinal smooth signaling and promoting extracellular matrix production. Devel-
muscle development. J Pediatr Gastroenterol Nutr 1996;23: opment 2006;133:833– 843.
379 –394. 47. Sandmann T, Jensen LJ, Jakobsen JS, et al. A temporal map of
27. Wallace KN, Akhter S, Smith EM, et al. Intestinal growth and transcription factor activity: mef2 directly regulates target genes
differentiation in zebrafish. Mech Dev 2005;122:157–173. at all stages of muscle development. Dev Cell 2006;10:797–
28. Andrew A. The origin of intramural ganglia. II. The trunk neural 807.
crest as a source of enteric ganglion cells. J Anat 1969;105: 48. Krauss S, Concordet JP, Ingham PW. A functionally conserved
89 –101. homolog of the Drosophila segment polarity gene hh is ex-
29. Cochard P, Le Douarin NM. Development of the intrinsic inner- pressed in tissues with polarizing activity in zebrafish embryos.
vation of the gut. Scand J Gastroenterol Suppl 1982;71:1–14. Cell 1993;75:1431–1444.
30. Gershon MD, Payette RF, Rothman TP. Development of the 49. Sakiyama J, Yokouchi Y, Kuroiwa A. HoxA and HoxB cluster
enteric nervous system. Fed Proc 1983;42:1620 –1625. genes subdivide the digestive tract into morphological domains
31. Rothman TP, Nilaver G, Gershon MD. Colonization of the devel- during chick development. Mech Dev 2001;101:233–236.
oping murine enteric nervous system and subsequent pheno- 50. Sakiyama J, Yokouchi Y, Kuroiwa A. Coordinated expression of
Hoxb genes and signaling molecules during development of the
typic expression by the precursors of peptidergic neurons.
chick respiratory tract. Dev Biol 2000;227:12–27.
J Comp Neurol 1984;225:13–23.
51. Grapin-Botton A, Melton DA. Endoderm development: from pat-
32. Henning SJ, Rubin DC, Shulman RJ. Ontogeny of the intestinal
terning to organogenesis. Trends Genet 2000;16:124 –130.
mucosa. Raven Press, 1994.
52. Beck F, Tata F, Chawengsaksophak K. Homeobox genes and gut
33. Wells JM, Melton DA. Early mouse endoderm is patterned by
development. Bioessays 2000;22:431– 441.
soluble factors from adjacent germ layers. Development 2000;
53. Yokouchi Y, Sakiyama J, Kuroiwa A. Coordinated expression of
127:1563–1572.
Abd-B subfamily genes of the HoxA cluster in the developing
34. Dessimoz J, Opoka R, Kordich JJ, et al. FGF signaling is neces-
digestive tract of chick embryo. Dev Biol 1995;169:76 – 89.
sary for establishing gut tube domains along the anterior-pos-
54. Stanfel MN, Moses KA, Schwartz RJ, et al. Regulation of organ
terior axis in vivo. Mech Dev 2006;123:42–55.
development by the NKX-homeodomain factors: an NKX code.
35. Sukegawa A, Narita T, Kameda T, et al. The concentric structure
Cell Mol Biol (Noisy-le-grand) 2005;Suppl 51:OL785–OL799.
of the developing gut is regulated by Sonic hedgehog derived
55. Park M, Lewis C, Turbay D, et al. Differential rescue of visceral
from endodermal epithelium. Development 2000;127:1971–
and cardiac defects in Drosophila by vertebrate tinman-related
1980.
genes. Proc Natl Acad Sci U S A 1998;95:9366 –9371.
36. Tseng HT, Shah R, Jamrich M. Function and regulation of FoxF1
56. Zakany J, Duboule D. Hox genes and the making of sphincters.
during Xenopus gut development. Development 2004;131:3637– Nature 1999;401:761–762.
3647. 57. Zacchetti G, Duboule D, Zakany J. Hox gene function in verte-
37. Zaffran S, Kuchler A, Lee HH, et al. biniou (FoxF), a central brate gut morphogenesis: the case of the caecum. Develop-
component in a regulatory network controlling visceral meso- ment 2007;134:3967–3973.
derm development and midgut morphogenesis in Drosophila. 58. Warot X, Fromental-Ramain C, Fraulob V, et al. Gene dosage-
Genes Dev 2001;15:2900 –2915. dependent effects of the Hoxa-13 and Hoxd-13 mutations on
38. Mahlapuu M, Enerback S, Carlsson P. Haploinsufficiency of the morphogenesis of the terminal parts of the digestive and uro-
forkhead gene Foxf1, a target for sonic hedgehog signaling, genital tracts. Development 1997;124:4781– 4791.
causes lung and foregut malformations. Development 2001; 59. Goodman FR, Bacchelli C, Brady AF, et al. Novel HOXA13 mu-
128:2397–2406. tations and the phenotypic spectrum of hand-foot-genital syn-
39. Jakobsen JS, Braun M, Astorga J, et al. Temporal ChIP-on-chip drome. Am J Hum Genet 2000;67:197–202.
reveals Biniou as a universal regulator of the visceral muscle 60. Goodman FR, Scambler PJ. Human HOX gene mutations. Clin
transcriptional network. Genes Dev 2007;21:2448 –2460. Genet 2001;59:1–11.
40. Mahlapuu M, Pelto-Huikko M, Aitola M, et al. FREAC-1 contains 61. Tennyson VM, Gershon MD, Sherman DL, et al. Structural ab-
a cell-type-specific transcriptional activation domain and is ex- normalities associated with congenital megacolon in transgenic
pressed in epithelial-mesenchymal interfaces. Dev Biol 1998; mice that overexpress the Hoxa-4 gene. Dev Dyn 1993;198:
202:183–195. 28 –53.
41. Mahlapuu M, Ormestad M, Enerback S, et al. The forkhead 62. Aubin J, Dery U, Lemieux M, et al. Stomach regional specifica-
transcription factor Foxf1 is required for differentiation of extra- tion requires Hoxa5-driven mesenchymal-epithelial signaling.
embryonic and lateral plate mesoderm. Development 2001; Development 2002;129:4075– 4087.
128:155–166. 63. Wang Z, Dolle P, Cardoso WV, et al. Retinoic acid regulates
42. Astorga J, Carlsson P. Hedgehog induction of murine vasculo- morphogenesis and patterning of posterior foregut derivatives.
genesis is mediated by Foxf1 and Bmp4. Development 2007; Dev Biol 2006;297:433– 445.
134:3753–3761. 64. Kumar M, Jordan N, Melton D, et al. Signals from lateral plate
43. Kalinichenko VV, Zhou Y, Bhattacharyya D, et al. Haploinsuffi- mesoderm instruct endoderm toward a pancreatic fate. Dev Biol
ciency of the mouse Forkhead Box f1 gene causes defects in 2003;259:109 –122.
2088 MCLIN ET AL GASTROENTEROLOGY Vol. 136, No. 7
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

65. Ryan JF, Baxevanis AD. Hox, Wnt, and the evolution of the 86. Smith DM, Nielsen C, Tabin CJ, et al. Roles of BMP signaling
primary body axis: insights from the early-divergent phyla. Biol and Nkx2.5 in patterning at the chick midgut-foregut boundary.
Direct 2007;2:37. Development 2000;127:3671–3681.
66. Lohnes D. The Cdx1 homeodomain protein: an integrator of 87. Henning SJ, Rubin DC, Shulman RJ. Ontogeny of the intestinal
posterior signaling in the mouse. Bioessays 2003;25:971– mucosa. In: LR J, ed. Physiology of the gastrointestinal tract.
980. New York, NY: Raven Press, 1994:571– 610.
67. Pilon N, Oh K, Sylvestre JR, et al. Cdx4 is a direct target of the 88. Lacroix B, Kedinger M, Simon-Assmann PM, et al. Effects of
canonical Wnt pathway. Dev Biol 2006;289:55– 63. human fetal gastroenteric mesenchymal cells on some devel-
68. Pilon N, Oh K, Sylvestre JR, et al. Wnt signaling is a key mediator opmental aspects of animal gut endoderm. Differentiation
of Cdx1 expression in vivo. Development 2007;134:2315– 1984;28:129 –135.
2323. 89. Ratineau C, Duluc I, Pourreyron C, et al. Endoderm- and mes-
69. Deschamps J, van Nes J. Developmental regulation of the Hox enchyme-dependent commitment of the differentiated epithelial
genes during axial morphogenesis in the mouse. Development cell types in the developing intestine of rat. Differentiation
2005;132:2931– 42. 2003;71:163–169.
70. Mandhan P, Quan QB, Beasley S, et al. Sonic hedgehog, BMP4, 90. Hayashi K, Yasugi S, Mizuno T. Pepsinogen gene transcription
and Hox genes in the development of anorectal malformations induced in heterologous epithelial-mesenchymal recombina-
in Ethylenethiourea-exposed fetal rats. J Pediatr Surg 2006;41: tions of chicken endoderms and glandular stomach mesen-
2041–2045. chyme. Development 1988;103:725–731.
71. Ramalho-Santos M, Melton DA, McMahon AP. Hedgehog signals 91. Rankin CT, Bunton T, Lawler AM, et al. Regulation of left-right
regulate multiple aspects of gastrointestinal development. De- patterning in mice by growth/differentiation factor-1. Nat Genet
velopment 2000;127:2763–2772. 2000;24:262–265.
72. Madison BB, McKenna LB, Dolson D, et al. FoxF1 and FoxL1 link 92. Collignon J, Varlet I, Robertson EJ. Relationship between asym-
hedgehog signaling and the control of epithelial proliferation in metric nodal expression and the direction of embryonic turning.
the developing stomach and intestine. J Biol Chem 2009;284: Nature 1996;381:155–158.
5936 –5944. 93. Sampath K, Cheng AM, Frisch A, et al. Functional differences
among Xenopus nodal-related genes in left-right axis determina-
73. Iafolla AK, Kahler SG. Megalencephaly in the neonatal period as
tion. Development 1997;124:3293–3302.
the initial manifestation of glutaric aciduria type I. J Pediatr
94. Yashiro K, Saijoh Y, Sakuma R, et al. Distinct transcriptional
1989;114:1004 –1006.
regulation and phylogenetic divergence of human LEFTY genes.
74. Piekarski DH, Stephens FD. The association and embryogene-
Genes Cells 2000;5:343–357.
sis of tracheo-oesophageal and anorectal anomalies. Prog Pe-
95. Logan M, Pagan-Westphal SM, Smith DM, et al. The transcrip-
diatr Surg 1976;9:63–76.
tion factor Pitx2 mediates situs-specific morphogenesis in res-
75. Nielsen C, Murtaugh LC, Chyung JC, et al. Gizzard formation and
ponse to left-right asymmetric signals. Cell 1998;94:
the role of Bapx1. Dev Biol 2001;231:164 –174.
307–317.
76. Yasugi S, Matsushita S, Mizuno T. Gland formation induced in
96. Yoshioka H, Meno C, Koshiba K, et al. Pitx2, a bicoid-type
the allantoic and small-intestinal endoderm by the proventricu-
homeobox gene, is involved in a lefty-signaling pathway in de-
lar mesenchyme is not coupled with pepsinogen expression.
termination of left-right asymmetry. Cell 1998;94:299 –305.
Differentiation 1985;30:47–52.
97. Davis NM, Kurpios NA, Sun X, et al. The chirality of gut rotation
77. Kedinger M, Simon-Assmann P, Bouziges F, et al. Epithelial-
derives from left-right asymmetric changes in the architecture of
mesenchymal interactions in intestinal epithelial differentiation.
the dorsal mesentery. Dev Cell 2008;15:134 –145.
Scand J Gastroenterol Suppl 1988;151:62– 69. 98. Lipscomb K, Schmitt C, Sablyak A, et al. Role for retinoid
78. Haffen K, Kedinger M, Simon-Assmann PM, et al. Mesenchyme- signaling in left-right asymmetric digestive organ morphogene-
dependent differentiation of intestinal brush-border enzymes in sis. Dev Dyn 2006;235:2266 –2275.
the gizzard endoderm of the chick embryo. Prog Clin Biol Res 99. El-Mounayri O, Triplett JW, Yates CW, et al. Regulation of
1982;85:261–270. smooth muscle-specific gene expression by homeodomain pro-
79. Ishizuya-Oka A, Mizuno T. Intestinal cytodifferentiation in vitro of teins, Hoxa10 and Hoxb8. J Biol Chem 2005;280:25854 –
chick stomach endoderm induced by the duodenal mesen- 25863.
chyme. J Embryol Exp Morphol 1984;82:163–176. 100. Louvard D, Kedinger M, Hauri HP. The differentiating intestinal
80. Roberts DJ. Molecular mechanisms of development of the gas- epithelial cell: establishment and maintenance of functions
trointestinal tract. Dev Dyn 2000;219:109 –120. through interactions between cellular structures. Annu Rev Cell
81. Goldstein AM, Brewer KC, Doyle AM, et al. BMP signaling is Biol 1992;8:157–195.
necessary for neural crest cell migration and ganglion formation 101. Kedinger M, Simon-Assmann PM, Lacroix B, et al. Fetal gut
in the enteric nervous system. Mech Dev 2005;122:821– 833. mesenchyme induces differentiation of cultured intestinal
82. Verzi MP, Stanfel MN, Moses KA, et al. Role of the homeodo- endodermal and crypt cells. Dev Biol 1986;113:474 – 483.
main transcription factor Bapx1 in mouse distal stomach devel- 102. Wang LC, Nassir F, Liu ZY, et al. Disruption of hedgehog signal-
opment. Gastroenterology 2009 Jan 14 [Epub ahead of print]. ing reveals a novel role in intestinal morphogenesis and intes-
83. Kim BM, Buchner G, Miletich I, et al. The stomach mesenchymal tinal-specific lipid metabolism in mice. Gastroenterology 2002;
transcription factor Barx1 specifies gastric epithelial identity 122:469 – 482.
through inhibition of transient Wnt signaling. Dev Cell 2005;8: 103. Madison BB, Braunstein K, Kuizon E, et al. Epithelial hedgehog
611– 622. signals pattern the intestinal crypt-villus axis. Development
84. Moniot B, Biau S, Faure S, et al. SOX9 specifies the pyloric 2005;132:279 –289.
sphincter epithelium through mesenchymal-epithelial signals. 104. Mishina Y. Function of bone morphogenetic protein signaling
Development 2004;131:3795–3804. during mouse development. Front Biosci 2003;8:d855– d869.
85. Theodosiou NA, Tabin CJ. Sox9 and Nkx2.5 determine the py- 105. Faure S, de Santa Barbara P, Roberts DJ, et al. Endogenous
loric sphincter epithelium under the control of BMP signaling. patterns of BMP signaling during early chick development. Dev
Dev Biol 2005;279:481– 490. Biol 2002;244:44 – 65.
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2089

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
106. De Santa Barbara P, Williams J, Goldstein AM, et al. Bone 128. Perreault N, Sackett SD, Katz JP, et al. Foxl1 is a mesenchymal
morphogenetic protein signaling pathway plays multiple roles Modifier of Min in carcinogenesis of stomach and colon. Genes
during gastrointestinal tract development. Dev Dyn 2005;234: Dev 2005;19:311–315.
312–322. 129. Katz JP, Perreault N, Goldstein BG, et al. Foxl1 null mice have
107. Batts LE, Polk DB, Dubois RN, et al. Bmp signaling is required abnormal intestinal epithelia, postnatal growth retardation, and
for intestinal growth and morphogenesis. Dev Dyn 2006;235: defective intestinal glucose uptake. Am J Physiol Gastrointest
1563–1570. Liver Physiol 2004;287:G856 –G864.
108. He XC, Zhang J, Tong WG, et al. BMP signaling inhibits intestinal 130. Oka T, Shiojima I, Monzen K, et al. Fibroblast growth factor
stem cell self-renewal through suppression of Wnt-beta-catenin plays a critical role in SM22alpha expression during Xenopus
signaling. Nat Genet 2004;36:1117–1121. embryogenesis. Arterioscler Thromb Vasc Biol 2000;20:907–
109. Auclair BA, Benoit YD, Rivard N, et al. Bone morphogenetic 914.
protein signaling is essential for terminal differentiation of the 131. Saint-Jeannet JP, Thiery JP, Koteliansky VE. Effect of an inhibi-
intestinal secretory cell lineage. Gastroenterology 2007;133: tory mutant of the FGF receptor on mesoderm-derived alpha-
smooth muscle actin-expressing cells in Xenopus embryo. Dev
887– 896.
Biol 1994;164:374 –382.
110. Haramis AP, Begthel H, van den Born M, et al. De novo crypt
132. Li X, Madison BB, Zacharias W, et al. Deconvoluting the intes-
formation and juvenile polyposis on BMP inhibition in mouse
tine: molecular evidence for a major role of the mesenchyme in
intestine. Science 2004;303:1684 –1686.
the modulation of signaling cross talk. Physiol Genomics 2007;
111. Ishizuya-Oka A, Hasebe T, Shimizu K, et al. Shh/BMP-4 signal-
29:290 –301.
ing pathway is essential for intestinal epithelial development
133. Vidrich A, Buzan JM, Ilo C, et al. Fibroblast growth factor recep-
during Xenopus larval-to-adult remodeling. Dev Dyn 2006;235: tor-3 is expressed in undifferentiated intestinal epithelial cells
3240 –3249. during murine crypt morphogenesis. Dev Dyn 2004;230:
112. Ishizuya-Oka A, Ueda S, Amano T, et al. Thyroid-hormone-depen- 114 –123.
dent and fibroblast-specific expression of BMP-4 correlates with 134. Zhang X, Stappenbeck TS, White AC, et al. Reciprocal epithelial-
adult epithelial development during amphibian intestinal remod- mesenchymal FGF signaling is required for cecal development.
eling. Cell Tissue Res 2001;303:187–195. Development 2006;133:173–180.
113. Pinto D, Clevers H. Wnt control of stem cells and differentiation 135. Shulman RJ, Schanler RJ, Lau C, et al. Early feeding, antenatal
in the intestinal epithelium. Exp Cell Res 2005;306:357–363. glucocorticoids, and human milk decrease intestinal permeabil-
114. Pinto D, Clevers H. Wnt, stem cells and cancer in the intestine. ity in preterm infants. Pediatr Res 1998;44:519 –523.
Biol Cell 2005;97:185–196. 136. Stallmach A, Hahn U, Merker HJ, et al. Differentiation of rat
115. Pinto D, Gregorieff A, Begthel H, et al. Canonical Wnt signals are intestinal epithelial cells is induced by organotypic mesenchy-
essential for homeostasis of the intestinal epithelium. Genes mal cells in vitro. Gut 1989;30:959 –970.
Dev 2003;17:1709 –1713. 137. Kedinger M, Simon-Assmann P, Alexandre E, et al. Importance
116. Clevers H. Wnt/beta-catenin signaling in development and dis- of a fibroblastic support for in vitro differentiation of intestinal
ease. Cell 2006;127:469 – 480. endodermal cells and for their response to glucocorticoids. Cell
117. van Es JH, Jay P, Gregorieff A, et al. Wnt signalling induces Differ 1987;20:171–182.
maturation of Paneth cells in intestinal crypts. Nat Cell Biol 138. Foltzer-Jourdainne C, Kedinger M, Raul F. Perinatal expression
2005;7:381–386. of brush-border hydrolases in rat colon: hormonal and tissue
118. Andoh A, Bamba S, Brittan M, et al. Role of intestinal subepi- regulations. Am J Physiol 1989;257:G496 –G503.
thelial myofibroblasts in inflammation and regenerative re- 139. Walsh MJ, LeLeiko NS, Sterling KM Jr. Regulation of types I, III,
sponse in the gut. Pharmacol Ther 2007;114:94 –106. and IV procollagen mRNA synthesis in glucocorticoid-mediated
119. Gregorieff A, Pinto D, Begthel H, et al. Expression pattern of Wnt intestinal development. J Biol Chem 1987;262:10814 –10818.
signaling components in the adult intestine. Gastroenterology 140. Simon-Assmann P, Kedinger M, De Arcangelis A, et al. Extracel-
2005;129:626 – 638. lular matrix components in intestinal development. Experientia
120. Theodosiou NA, Tabin CJ. Wnt signaling during development of 1995;51:883–900.
the gastrointestinal tract. Dev Biol 2003;259:258 –271. 141. Weiser MM, Sykes DE, Killen PD. Rat intestinal basement mem-
brane synthesis: Epithelial versus nonepithelial contributions.
121. Muncan V, Faro A, Haramis AP, et al. T-cell factor 4 (Tcf7l2)
Lab Invest 1990;62:325–330.
maintains proliferative compartments in zebrafish intestine.
142. Simo P, Simon-Assmann P, Arnold C, et al. Mesenchyme-medi-
EMBO Rep 2007;8:966 –973.
ated effect of dexamethasone on laminin in cocultures of em-
122. Barker N, Huls G, Korinek V, et al. Restricted high level expres-
bryonic gut epithelial cells and mesenchyme-derived cells. J Cell
sion of Tcf-4 protein in intestinal and mammary gland epithe-
Sci 1992;101:161–171.
lium. Am J Pathol 1999;154:29 –35.
143. Solomon NS, Gartner H, Oesterreicher TJ, Henning SJ. Devel-
123. Booth C, Brady G, Potten CS. Crowd control in the crypt. Nat
opment of glucocorticoid-responsiveness in mouse intestine.
Med 2002;8:1360 –1361. Pediatr Res 2001;49:782– 8.
124. Kim BM, Mao J, Taketo MM, et al. Phases of canonical Wnt 144. Gartner H, Graul MC, Oesterreicher TJ, et al. Development of
signaling during the development of mouse intestinal epithe- the fetal intestine in mice lacking the glucocorticoid receptor
lium. Gastroenterology 2007;133:529 –538. (GR). J Cell Physiol 2003;194:80 – 87.
125. Cervantes S, Yamaguchi TP, Hebrok M. Wnt5a is essential for 145. Fraichard A, Chassande O, Plateroti M, et al. The T3R alpha
intestinal elongation in mice. Dev Biol 2009;326:285–294. gene encoding a thyroid hormone receptor is essential for post-
126. Geske MJ, Zhang X, Patel KK, et al. Fgf9 signaling regulates natal development and thyroid hormone production. EMBO J
small intestinal elongation and mesenchymal development. De- 1997;16:4412– 4420.
velopment 2008;135:2959 –2968. 146. Schreiber AM, Cai L, Brown DD. Remodeling of the intestine
127. Kaestner KH, Silberg DG, Traber PG, et al. The mesenchymal during metamorphosis of Xenopus laevis. Proc Natl Acad Sci
winged helix transcription factor Fkh6 is required for the control U S A 2005;102:3720 –3725.
of gastrointestinal proliferation and differentiation. Genes Dev 147. Leeper LL, McDonald MC, Heath JP, et al. Sucrase-isomaltase
1997;11:1583–1595. ontogeny: synergism between glucocorticoids and thyroxine re-
2090 MCLIN ET AL GASTROENTEROLOGY Vol. 136, No. 7
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

flects increased mRNA and no change in cell migration. 168. Frasch M. Induction of visceral and cardiac mesoderm by ecto-
Biochem Biophys Res Commun 1998;246:765–770. dermal Dpp in the early Drosophila embryo. Nature 1995;374:
148. McDonald MC, Henning SJ. Synergistic effects of thyroxine and 464 – 467.
dexamethasone on enzyme ontogeny in rat small intestine. 169. Lee HH, Frasch M. Nuclear integration of positive Dpp signals,
Pediatr Res 1992;32:306 –311. antagonistic Wg inputs and mesodermal competence factors
149. Su Y, Shi Y, Shi YB. Cyclosporin A but not FK506 inhibits thyroid during Drosophila visceral mesoderm induction. Development
hormone-induced apoptosis in tadpole intestinal epithelium. 2005;132:1429 –1442.
FASEB J 1997;11:559 –565. 170. Stickney HL, Imai Y, Draper B, et al. Zebrafish bmp4 functions
150. Su Y, Shi Y, Stolow MA, et al. Thyroid hormone induces apopto- during late gastrulation to specify ventroposterior cell fates. Dev
sis in primary cell cultures of tadpole intestine: cell type spec- Biol 2007;310:71– 84.
ificity and effects of extracellular matrix. J Cell Biol 1997;139: 171. McMahon JA, Takada S, Zimmerman LB, et al. Noggin-mediated
1533–1543. antagonism of BMP signaling is required for growth and pattern-
151. Ishizuya-Oka A, Shi YB. Regulation of adult intestinal epithelial ing of the neural tube and somite. Genes Dev 1998;12:1438 –
stem cell development by thyroid hormone during Xenopus 1452.
laevis metamorphosis. Dev Dyn 2007;236:3358 –3368. 172. Monsoro-Burq AH, Duprez D, Watanabe Y, et al. The role of bone
152. Quasnichka H, Slater SC, Beeching CA, et al. Regulation of morphogenetic proteins in vertebral development. Development
smooth muscle cell proliferation by beta-catenin/T-cell factor 1996;122:3607–3616.
signaling involves modulation of cyclin D1 and p21 expression. 173. Xue L, Chen X, Chang Y, et al. Regulatory elements of the EKLF
Circ Res 2006;99:1329 –1337. gene that direct erythroid cell-specific expression during mam-
153. Karlsson L, Lindahl P, Heath JK, et al. Abnormal gastrointestinal malian development. Blood 2004;103:4078 – 4083.
development in PDGF-A and PDGFR-(alpha) deficient mice impli- 174. Davidson AJ, Zon LI. Turning mesoderm into blood: the forma-
cates a novel mesenchymal structure with putative instructive tion of hematopoietic stem cells during embryogenesis. Curr
properties in villus morphogenesis. Development 2000;127: Top Dev Biol 2000;50:45– 60.
3457–3466. 175. Dosch R, Gawantka V, Delius H, et al. Bmp-4 acts as a mor-
154. Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function muta- phogen in dorsoventral mesoderm patterning in Xenopus. De-
tions of c-kit in human gastrointestinal stromal tumors. Science velopment 1997;124:2325–2334.
1998;279:577–580. 176. Onichtchouk D, Glinka A, Niehrs C. Requirement for Xvent-1 and
155. Taniguchi M, Nishida T, Hirota S, et al. Effect of c-kit mutation
Xvent-2 gene function in dorsoventral patterning of Xenopus
on prognosis of gastrointestinal stromal tumors. Cancer Res
mesoderm. Development 1998;125:1447–1456.
1999;59:4297– 4300.
177. Ault KT, Dirksen ML, Jamrich M. A novel homeobox gene PV.1
156. Rubin BP. Gastrointestinal stromal tumours: an update. Histo-
mediates induction of ventral mesoderm in Xenopus embryos.
pathology 2006;48:83–96.
Proc Natl Acad Sci U S A 1996;93:6415– 6420.
157. Heinrich MC, Rubin BP, Longley BJ, et al. Biology and genetic
178. Onichtchouk D, Gawantka V, Dosch R, et al. The Xvent-2 ho-
aspects of gastrointestinal stromal tumors: KIT activation and
meobox gene is part of the BMP-4 signalling pathway controlling
cytogenetic alterations. Hum Pathol 2002;33:484 – 495.
[correction of controling] dorsoventral patterning of Xenopus
158. Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating
mesoderm. Development 1996;122:3045–3053.
mutations in gastrointestinal stromal tumors. Science 2003;
179. Kozmik Z, Holland LZ, Schubert M, et al. Characterization of
299:708 –710.
Amphioxus AmphiVent, an evolutionarily conserved marker for
159. MacDonald RS. The role of insulin-like growth factors in small
chordate ventral mesoderm. Genesis 2001;29:172–179.
intestinal cell growth and development. Horm Metab Res 1999;
180. Bienz M, Tremml G. Domain of Ultrabithorax expression in
31:103–113.
Drosophila visceral mesoderm from autoregulation and exclu-
160. Michaylira CZ, Simmons JG, Ramocki NM, et al. Suppressor of
cytokine signaling-2 limits intestinal growth and enterotrophic sion. Nature 1988;333:576 –578.
actions of IGF-I in vivo. Am J Physiol Gastrointest Liver Physiol 181. Jagla K, Bellard M, Frasch M. A cluster of Drosophila homeobox
2006;291:G472–G481. genes involved in mesoderm differentiation programs. Bioes-
161. Fukamachi H, Narita T, Yahagi N, et al. Endothelin-3 controls says 2001;23:125–133.
growth of colonic epithelial cells by mediating epithelial-mesen- 182. Azpiazu N, Frasch M. tinman and bagpipe: two homeo box
chymal interaction. Dev Growth Differ 2005;47:573–580. genes that determine cell fates in the dorsal mesoderm of
162. Wang Y, Wang L, Iordanov H, et al. Epimorphin(⫺/⫺) mice have Drosophila. Genes Dev 1993;7:1325–1340.
increased intestinal growth, decreased susceptibility to dextran 183. Azpiazu N, Lawrence PA, Vincent JP, et al. Segmentation and
sodium sulfate colitis, and impaired spermatogenesis. J Clin specification of the Drosophila mesoderm. Genes Dev 1996;
Invest 2006;116:1535–1546. 10:3183–3194.
163. Hirai Y, Takebe K, Takashina M, et al. Epimorphin: a mesen- 184. Lo PC, Frasch M. bagpipe-Dependent expression of vimar, a
chymal protein essential for epithelial morphogenesis. Cell novel Armadillo-repeats gene, in Drosophila visceral mesoderm.
1992;69:471– 481. Mech Dev 1998;72:65–75.
164. Wallace KN, Dolan AC, Seiler C, et al. Mutation of smooth 185. Newman CS, Grow MW, Cleaver O, et al. Xbap, a vertebrate
muscle myosin causes epithelial invasion and cystic expansion gene related to bagpipe, is expressed in developing craniofacial
of the zebrafish intestine. Dev Cell 2005;8:717–726. structures and in anterior gut muscle. Dev Biol 1997;181:223–
165. Timpl R, Brown JC. Supramolecular assembly of basement 233.
membranes. Bioessays 1996;18:123–132. 186. Newman CS, Krieg PA. The Xenopus bagpipe-related homeobox
166. Shi YB, Fu L, Hasebe T, et al. Regulation of extracellular matrix gene zampogna is expressed in the pharyngeal endoderm and
remodeling and cell fate determination by matrix metalloprotein- the visceral musculature of the midgut. Dev Genes Evol 1999;
ase stromelysin-3 during thyroid hormone-dependent post-em- 209:132–134.
bryonic development. Pharmacol Ther 2007;116:391– 400. 187. Nieuwkoop D, Faber J. Normal table of Xenopus laevis (Daudin).
167. El-Hodiri H, Bhatia-Dey N, Kenyon K, et al. Fox (forkhead) genes New York, NY: Garland Publishing Inc, 1994.
are involved in the dorso-ventral patterning of the Xenopus 188. Horb ME. Patterning the endoderm: the importance of neigh-
mesoderm. Int J Dev Biol 2001;45:265–271. bours. Bioessays 2000;22:599 – 602.
June 2009 VISCERAL MESODERM IN GI DEVELOPMENT 2091

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
189. Horb ME, Slack JM. Endoderm specification and differentiation sophila homeobox gene tinman: differential expression of cNKx-
in Xenopus embryos. Dev Biol 2001;236:330 –343. 2.3 and cNKx-2.5 during heart and gut development. Mech Dev
190. Swindell EC, Eichele G. Retinoid metabolizing enzymes in de- 1996;56:151–163.
velopment. Biofactors 1999;10:85– 89.
191. McLin VA, Rankin SA, Zorn AM. Repression of Wnt/beta-catenin
signaling in the anterior endoderm is essential for liver and
Received September 19, 2008. Accepted March 4, 2009.
pancreas development. Development 2007;134:2207–2217.
192. Glinka A, Wu W, Onichtchouk D, et al. Head induction by simul-
taneous repression of Bmp and Wnt signalling in Xenopus. Reprint requests
Nature 1997;389:517–519. Address requests for reprints to: Valérie A. McLin, MD, Unite de
193. Okubo T, Hogan BL. Hyperactive Wnt signaling changes the devel- Gastroenterologie, Hepatologie et Nutrition Pediatrique, Hopital des
opmental potential of embryonic lung endoderm. J Biol 2004;3:11. Enfants, CH-1211 Geneva, Switzerland. e-mail: valerie.mclin@
194. Crosnier C, Stamataki D, Lewis J. Organizing cell renewal in the hcuge.ch.
intestine: stem cells, signals and combinatorial control. Nat Rev
Genet 2006;7:349 –359. Acknowledgments
195. Lints TJ, Hartley L, Parsons LM, et al. Mesoderm-specific ex- The authors thank all the reviewers for their encouragement
pression of the divergent homeobox gene Hlx during murine and in particular reviewers 1 and 3 for extremely constructive
embryogenesis. Dev Dyn 1996;205:457– 470. suggestions. V.A.M. thanks Peter M. Carson for enduring
196. Bates MD, Schatzman LC, Lints T, et al. Structural and func- support.
tional characterization of the mouse Hlx homeobox gene.
Mamm Genome 2000;11:836 – 842. Conflicts of interest
197. Hentsch B, Lyons I, Li R, et al. Hlx homeo box gene is essential The authors disclose no conflicts.
for an inductive tissue interaction that drives expansion of
embryonic liver and gut. Genes Dev 1996;10:70 –79. Funding
198. Pabst O, Schneider A, Brand T, et al. The mouse Nkx2-3 home- V.A.M. is supported by the National Institutes of Health
odomain gene is expressed in gut mesenchyme during pre- and (K08DK078656) and a Young Investigator Award from the
postnatal mouse development. Dev Dyn 1997;209:29 –35. Children’s Digestive Health and Nutrition Foundation, S.J.H. is
199. Buchberger A, Pabst O, Brand T, et al. Chick NKx-2.3 represents supported by the National Institutes of Health (R01DK069585), and
a novel family member of vertebrate homologues to the Dro- M.J. is supported by the Retinal Research Foundation.

View publication stats

You might also like