New SnapShot: The Intestinal Crypt - Cell · 2015. 1. 30. · The proximal region, the small...

3
Cdx2 Proximodistal organization Gata4 Represses Ileal gene program Differentiated cell compartment Transient-amplifying compartment Stem cell niche Duodenum Jejunum Ileum Prox. colon Dist. colon Caecum Represses stomach gene program M cell Enterocyte Goblet Endocrine NeuroD Elf3 Cdx2+Hnf4 Elf3 Spdef Pax4/6 Nkx2.2 Gfi1 RankL Notch Dll1/4 Absorptive progenitor Secretory progenitor Gfi1 Sox9 Spdef Math1 +WNT Damage Damage Paneth cell ISCs (Lgr5+) Spi-B Ngn3 ? ? Tuft LRC (Lgr5+) Hes1 Math1 Cell hierarchy and lineage specification TIME Restricted niche for stem cells Intestinal stem cells at the crypt base Neutral competition in the ISC niche Paneth cells Lgr5+ stem cells Symmetric stem cell divisions Growth of surviving clones and extinction of neighboring clones Crypts inevitably drift toward clonality Anoikis Nutrients Cell amplification Migration PEYER’S PATCH CRYPT VILLUS Lymphoid cells Lymphoid cells Enterocytes Nutrients absorption Transient amplifying cells Proliferation Lineage commitment Paneth cells Innate immune response Stem cell niche maintenance Tuft cells Opioids release Prostanoids production Other? Goblet cells Mucus secretion Enteroendocrine cells Hormone production M cells Antigen sampling Intestinal stem cells (ISCs) Tissue renewal Label retaining cells (LRCs) Paneth cell precursors Quiescent cells Regeneration of ISCs The small intestine WNT WNT WNT WNT RSPO β-cat β-cat NOTCH NOTCH-IC DLL1/4 LRIG1 PANETH CELL ISC (CBC) ERBB Proliferation Inhibition of the secretory program: HES1 Cell positioning: EPHB2 Silencing of cell-cycle inhibitors (p27kip, p57kip) Repression of expression of Dll1/4 Proliferation, block differentiation: MYC Regulation of WNT signaling: ZNRF3/RNF43, LGR5 Cell positioning: EPHB2, EPHB3 Self-renewal: ASCL2 BMPRI ZNRF3/ RNF43 ? ? ? STROMAL CELL GREM BMP LGR4/5 FRIZZLED LRP6 TCF CSL BMPRII TGFα EGF Signaling in ISCs See online version for legend and references. 1198 Cell 152, February 28, 2013 ©2013 Elsevier Inc. DOI http://dx.doi.org/10.1016/j.cell.2013.02.030 SnapShot: The Intestinal Crypt Hans Clevers 1 and Eduard Batlle 2,3 1 Hubrecht Institute, KNAW and University Medical Centre Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands 2 Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain 3 Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain

Transcript of New SnapShot: The Intestinal Crypt - Cell · 2015. 1. 30. · The proximal region, the small...

Page 1: New SnapShot: The Intestinal Crypt - Cell · 2015. 1. 30. · The proximal region, the small intestine (duodenum, jejunum, and ileum), is arranged in invaginations (crypts) intercalated

Cd

x2

Pro

xim

od

ista

l org

an

iza

tio

n

Gat

a4R

epre

sses

Ile

al g

ene

pro

gra

m

Diff

eren

tiat

ed c

ell

com

par

tmen

t

Tran

sien

t-am

plif

ying

com

par

tmen

t

Ste

m c

ell n

iche

Duo

den

umJe

junu

mIle

umP

rox.

co

lon

Dis

t. c

olo

n

Cae

cum

Rep

ress

es s

tom

ach

gen

e p

rog

ram

gen

e p

rog

ram

Sto

mac

h

M c

ell

En

tero

cyte

Go

ble

tE

nd

ocr

ine

Neu

roD

Elf3

Cd

x2+

Hnf

4

Elf3

Sp

def

Pax

4/6

Nkx

2.2

G�1

Ran

kL

No

tch

Dll1

/4A

bso

rpti

vep

rog

enit

or

Sec

reto

ry p

rog

enit

or

G�1

So

x9S

pd

efM

ath1

+W

NT

Dam

age

Dam

age

Pan

eth

cel

lIS

Cs

(Lg

r5+

)

Sp

i-B

Ng

n3?

?

Tuft

LR

C(L

gr5

+)

Hes

1M

ath1

Ce

ll h

iera

rch

y a

nd

lin

ea

ge

sp

ec

ific

ati

on

TIM

E

Res

tric

ted

nic

hefo

r st

em c

ells

Inte

stin

al s

tem

cel

ls a

t th

e cr

ypt

bas

e

Ne

utr

al c

om

pe

titi

on

inth

e I

SC

nic

he

Pan

eth

cells

Lgr5

+ s

tem

cel

ls

Sym

met

ric

stem

cell

div

isio

ns

Gro

wth

of

surv

ivin

g

clo

nes

and

ext

inct

ion

of

neig

hbo

ring

clo

nes

Cry

pts

inev

itab

ly d

rift

tow

ard

clo

nalit

y

An

oik

is

Nut

rien

ts

Ce

ll a

mp

lific

ati

on

Mig

rati

on

PE

YE

R’S

PA

TC

H

CR

YP

T

VIL

LU

S

Lym

ph

oid

cel

lsLy

mp

ho

id c

ells

En

tero

cyte

sN

utri

ents

ab

sorp

tio

n

Tran

sien

t am

plif

yin

g c

ells

Pro

lifer

atio

nLi

neag

e co

mm

itm

ent

Pan

eth

cel

lsIn

nate

imm

une

resp

ons

eS

tem

cel

l nic

he m

aint

enan

ce

Tuft

cel

lsO

pio

ids

rele

ase

Pro

stan

oid

s p

rod

ucti

on

Oth

er?

Go

ble

t ce

llsM

ucus

sec

reti

on

En

tero

end

ocr

ine

cells

Ho

rmo

ne p

rod

ucti

on

M c

ells

Ant

igen

sam

plin

g

Inte

stin

al s

tem

cel

ls (

ISC

s)T

issu

e re

new

al

Lab

el r

etai

nin

g c

ells

(L

RC

s)P

anet

h ce

ll p

recu

rso

rsQ

uies

cent

cel

lsR

egen

erat

ion

of

ISC

s

Th

e s

ma

ll in

test

ine

WN

TW

NT

WN

T

WN

T

RS

PO

β-ca

t

β-ca

t

NO

TC

HN

OT

CH

-IC

DLL

1/4

LRIG

1

PA

NE

TH

CE

LL

ISC

(C

BC

)

ER

BB

Pro

lifer

atio

n

Inhi

bit

ion

of

the

secr

eto

ry p

rog

ram

: H

ES

1C

ell p

osi

tio

ning

: E

PH

B2

Sile

ncin

g o

f ce

ll-cy

cle

inhi

bit

ors

(p

27ki

p,

p57

kip

)R

epre

ssio

n o

f ex

pre

ssio

n o

f D

ll1/4

Pro

lifer

atio

n, b

lock

diff

eren

tiat

ion:

MY

CR

egul

atio

n o

f W

NT

sig

nalin

g:

ZN

RF

3/R

NF

43,

LGR

5C

ell p

osi

tio

ning

: E

PH

B2,

EP

HB

3S

elf-

rene

wal

: A

SC

L2

BM

PR

I

ZN

RF

3/R

NF

43

?

?

?

ST

RO

MA

L C

EL

L

GR

EM

BM

P

LGR

4/5

FR

IZZ

LED

LRP

6

TC

F

CS

L

BM

PR

II

TG

EG

F

Sig

na

ling

in I

SC

s

See online version for legend and references.1198 Cell 152, February 28, 2013 ©2013 Elsevier Inc. DOI http://dx.doi.org/10.1016/j.cell.2013.02.030

Snap

Shot:

The Inte

stin

al C

rypt

Han

s C

leve

rs1

and

Ed

uard

Bat

lle2,

3

1 Hub

rech

t In

stitu

te, K

NA

W a

nd U

nive

rsity

Med

ical

Cen

tre

Utr

echt

, Up

psa

lala

an 8

, 358

4CT

Utr

echt

, the

Net

herla

nds

2 Ins

titut

e fo

r R

esea

rch

in B

iom

edic

ine

(IRB

Bar

celo

na),

0802

8 B

arce

lona

, Sp

ain

3 Ins

tituc

ió C

atal

ana

de

Rec

erca

i E

stud

is A

vanç

ats

(ICR

EA

), 08

010

Bar

celo

na, S

pai

n

Page 2: New SnapShot: The Intestinal Crypt - Cell · 2015. 1. 30. · The proximal region, the small intestine (duodenum, jejunum, and ileum), is arranged in invaginations (crypts) intercalated

1198.e1 Cell 152, February 28, 2013 ©2013 Elsevier Inc. DOI http://dx.doi.org/10.1016/j.cell.2013.02.030

SnapShot: The Intestinal CryptHans Clevers1 and Eduard Batlle2,3

1Hubrecht Institute, KNAW and University Medical Centre Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands2Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain3Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain

Organization of the Small Intestine and the ColonThe inner surface of the intestinal tube is lined by a simple epithelium, which displays distinct morphologies along the proximo-distal axis. The proximal region, the small intestine (duodenum, jejunum, and ileum), is arranged in invaginations (crypts) intercalated with finger-like protrusions (villus) that represent units specialized in the absorption of micronu-trients. The colon consists only of crypts and is largely dedicated to the compaction of stool. The identity of each of these segments is, in part, specified through the expression of the homeotic transcription factor Cdx2, which represses the expression of genes characteristic of the stomach, and the zinc finger Gata4, which confers proximal identity to the duodenum and ileum. Despite their distinctive morphologies, epithelial cell types of both the small intestine and colon are organized following a bottom-to-top axis into three compartments: the stem cell compartment that is located at the crypt base, the transient-amplifying (TA) compartment that occupies the middle portion of the crypts, and the differentiation zone, which expands from the top third of the crypt and the surface epithelium to the tip of the villus.

Renewal of the Epithelial Layer and Lineage SpecificationSix differentiated cell types populate the intestinal epithelium. The most abundant cells are the enterocytes (i.e., adsorptive cells) and goblet cells (i.e., mucosecreting cells). Other rare secretory cell types are scattered throughout the epithelium; they are enteroendocrine cells, which produce a diverse array of hormones, and the tuft cells, which are believed to secrete prostanoids. Microfold cells (M cells) are specialized epithelial cells situated over Peyer’s patches (PP) that transport antigens into intraepithelial pockets accessed by antigen-presenting cells. Finally, Paneth cells reside at the crypt base and perform a dual role; they secrete antimicrobial substances but also nurture the intestinal stem cell (ISC) population.

All differentiated cell types in the intestinal epithelium are short lived. Enterocytes, goblet cells, and the other secretory lineages are born in the crypts and follow an upward migratory flow that carries them to the tip of the villi in ~1 week. At this location, they are extruded into the lumen. Paneth cells are the exception and are retained at the base of the crypts, where they live for 6–8 weeks. Renewal of the epithelial layer is sustained throughout life by a small number of ISCs (n = 10–15 per crypt), which are located at the bottom-most positions intermingled with Paneth cells. ISCs proliferate with a rate of approximately one division per day. Their progeny is amplified through a series of very rapid divisions (about one division every 12 hr). While these TA cells migrate upward, they become progressively committed toward one of six lineages that are present in the intestine.

The activity of the bHLH transcription factor Math1 commits precursor cells to a secretory phenotype. Math1 also promotes elevated levels of delta-like ligands (Dll1/Dll4) in secretory precursors. In contrast, expression of Math1 is repressed in enterocytes by the Notch downstream effector Hes1. As a result of lateral inhibition, differential Notch activity in adjacent precursors operates as a binary switch to specify absorptive versus secretory cell types. Following commitment, a complex cascade of transcription factors drives the differentiation of the secretory precursor into distinct mature cell types (goblet, enteroendocrine, tuft, or Paneth cells). Differentiation of M cells requires the transcrip-tion factor Spi-B, the expression of which is switched on downstream of RANK signaling. RANK-L, the ligand for RANK receptor, is secreted by stromal cells present at Peyer’s patches.

Quiescent versus Proliferative ISCsIn homeostasis, Lgr5+ ISCs generate all cell types present in the epithelium (Barker et al., 2007; Sato et al., 2009). An independent class of quiescent ISCs marked by Bmi1, Lrig1, Tert, and Hopx has been proposed to serve as “reserve” stem cells. A recent study provides a simple view of the connection between these two stem cell types (Buczacki et al., 2013). The study shows that a small subset of Lgr5+ cells can enter a quiescent state. These label-retaining cells (LRCs) localize around crypt position +3. Lgr5+ LRCs express most of the markers previously postulated for the quiescent ISC population, but also several Paneth-cell-specific genes. These LRCs are not ISCs but, rather, transient precursors of Paneth cells, as they are short lived under homeostatic conditions (2–3 weeks) and they all differentiate toward mature Paneth cells. However, upon loss of the ISC pool, they can revert to become cycling Lrg5+ ISCs. Dedifferentiation of Dll1+-committed secretory precursors upon irradiation followed by regeneration of the ISC pool has also been demonstrated (van Es et al., 2012).

Neutral Competition in the ISC NicheAn intestinal crypt contains about 14 equal ISCs that all divide each day. Their dynamics are consistent with a model in which the resident ISCs double their numbers each day and stochastically adopt either stem or TA fates. Thus, ISCs divide symmetrically while competing for a niche of limited size. As a consequence, their turnover follows a pattern of neutral drift dynamics and crypts tend toward clonality within a period of 1–6 months. Therefore, ISCs persist for life as a population, yet only the lineage of one particular ISC is present in each crypt at any given time (Snippert et al., 2010).

Signaling in ISCsISCs are specified by high levels of WNT signaling in the crypts. R-spondin binds to LGR4/LGR5 receptors and potentiates WNT signals in ISCs (de Lau et al., 2011). On the contrary, the activity of Znfr3/Rnf43 negatively controls WNT signals in the ISC pool by ubiquitinating Frizzled receptors (Koo et al., 2012). Paneth cells secrete the WNT3 ligand constitutively, but an additional Wnt source also exists in the surrounding stroma. Notch signaling mainly acts by inhibiting the secretory fate in ISCs (Pellegrinet et al., 2011). Notch ligands (Dll1 and Dll4) are expressed by surrounding secretory cells, including the Paneth cells. Mitogenic stimuli regulate the size of the proliferative compartment. Lrig1, a marker for stem and early TA cells, acts as a negative regulator of receptor tyrosine kinase (RTK) activity (Wong et al., 2012). BMP signaling inhibits the stem cell fate in the intestine (Haramis et al., 2004) probably by antagonizing WNT signaling. BMPs are mainly expressed by stromal cells that surround the epithelium, whereas ISCs are protected from their action by the presence of local inhibitors, including Gremlin.

RefeRences

Barker, N., van Es, J.H., Kuipers, J., Kujala, P., van den Born, M., Cozijnsen, M., Haegebarth, A., Korving, J., Begthel, H., Peters, P.J., et al. (2007). Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007.

Buczacki, S.J.A., Zecchini, H.I., Nicholson, A.M., Russell, R., Vermeulen, L., Kemp, R., and Winton, D.J. (2013). The intestinal label-retaining cell expresses Lgr5 and is a committed secretory precursor. Nature. Published online February 27, 2013. http://dx.doi.org/10.1038/nature11965.

de Lau, W., Barker, N., Low, T.Y., Koo, B.K., Li, V.S., Teunissen, H., Kujala, P., Haegebarth, A., Peters, P.J., van de Wetering, M., et al. (2011). Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature 476, 293–297.

Haramis, A.P., Begthel, H., van den Born, M., van Es, J., Jonkheer, S., Offerhaus, G.J., and Clevers, H. (2004). De novo crypt formation and juvenile polyposis on BMP inhibition in mouse intestine. Science 303, 1684–1686.

Koo, B.K., Spit, M., Jordens, I., Low, T.Y., Stange, D.E., van de Wetering, M., van Es, J.H., Mohammed, S., Heck, A.J., Maurice, M.M., and Clevers, H. (2012). Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature 488, 665–669.

Page 3: New SnapShot: The Intestinal Crypt - Cell · 2015. 1. 30. · The proximal region, the small intestine (duodenum, jejunum, and ileum), is arranged in invaginations (crypts) intercalated

SnapShot: The Intestinal CryptHans Clevers1 and Eduard Batlle2,3

1Hubrecht Institute, KNAW and University Medical Centre Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands2Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain3Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain

Pellegrinet, L., Rodilla, V., Liu, Z., Chen, S., Koch, U., Espinosa, L., Kaestner, K.H., Kopan, R., Lewis, J., and Radtke, F. (2011). Dll1- and dll4-mediated notch signaling are required for homeostasis of intestinal stem cells. Gastroenterology 140, 1230–1240.e1–e7.

Sato, T., Vries, R.G., Snippert, H.J., van de Wetering, M., Barker, N., Stange, D.E., van Es, J.H., Abo, A., Kujala, P., Peters, P.J., and Clevers, H. (2009). Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265.

Snippert, H.J., van der Flier, L.G., Sato, T., van Es, J.H., van den Born, M., Kroon-Veenboer, C., Barker, N., Klein, A.M., van Rheenen, J., Simons, B.D., and Clevers, H. (2010). Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144.

van Es, J.H., Sato, T., van de Wetering, M., Lyubimova, A., Nee, A.N., Gregorieff, A., Sasaki, N., Zeinstra, L., van den Born, M., Korving, J., et al. (2012). Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat. Cell Biol. 14, 1099–1104.

Wong, V.W., Stange, D.E., Page, M.E., Buczacki, S., Wabik, A., Itami, S., van de Wetering, M., Poulsom, R., Wright, N.A., Trotter, M.W., et al. (2012). Lrig1 controls intestinal stem-cell homeostasis by negative regulation of ErbB signalling. Nat. Cell Biol. 14, 401–408.

1198.e2 Cell 152, February 28, 2013 ©2013 Elsevier Inc. DOI http://dx.doi.org/10.1016/j.cell.2013.02.030