ArticlePDF Available

Intestinal crypt homeostasis revealed at single-stem-cell level by in vivo live imaging

Authors:
  • Princess Máxima Center

Abstract and Figures

The rapid turnover of the mammalian intestinal epithelium is supported by stem cells located around the base of the crypt. In addition to the Lgr5 marker, intestinal stem cells have been associated with other markers that are expressed heterogeneously within the crypt base region. Previous quantitative clonal fate analyses have led to the proposal that homeostasis occurs as the consequence of neutral competition between dividing stem cells. However, the short-term behaviour of individual Lgr5(+) cells positioned at different locations within the crypt base compartment has not been resolved. Here we establish the short-term dynamics of intestinal stem cells using the novel approach of continuous intravital imaging of Lgr5-Confetti mice. We find that Lgr5(+) cells in the upper part of the niche (termed 'border cells') can be passively displaced into the transit-amplifying domain, after the division of proximate cells, implying that the determination of stem-cell fate can be uncoupled from division. Through quantitative analysis of individual clonal lineages, we show that stem cells at the crypt base, termed 'central cells', experience a survival advantage over border stem cells. However, through the transfer of stem cells between the border and central regions, all Lgr5(+) cells are endowed with long-term self-renewal potential. These findings establish a novel paradigm for stem-cell maintenance in which a dynamically heterogeneous cell population is able to function long term as a single stem-cell pool.
Content may be subject to copyright.
Intestinal crypt homeostasis revealed at single stem cell level by
in vivo live-imaging
Laila Ritsma#1,2, Saskia I.J. Ellenbroek#1,2, Anoek Zomer1,2, Hugo J. Snippert2,3, Frederic J.
de Sauvage4, Benjamin D. Simons5,6,7, Hans Clevers1,2, and Jacco van Rheenen1,2
1Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT, Utrecht,
the Netherlands 2Cancer Genomics Netherlands 3University Medical Centre Utrecht,
Universiteitsweg 100, 3584 CG, Utrecht, the Netherlands 4Department of Molecular Biology,
Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA 5Cavendish Laboratory,
Department of Physics, J.J. Thomson Avenue, University of Cambridge, Cambridge CB3 0HE,
UK. 6The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis
Court Road, Cambridge CB2 1QN, UK. 7Wellcome Trust-Medical Research Council Stem Cell
Institute, University of Cambridge, UK.
# These authors contributed equally to this work.
Summary
The rapid turnover of the mammalian intestinal epithelium is supported by stem cells located
around the base of the crypt1. Alongside Lgr5, intestinal stem cells have been associated with
various markers, which are expressed heterogeneously within the crypt base region1-6. Previous
quantitative clonal fate analyses have proposed that homeostasis occurs as the consequence of
neutral competition between dividing stem cells7-9. However, the short-term behaviour of
individual Lgr5+ cells positioned at different locations within the crypt base compartment has not
been resolved. Here, we established the short-term dynamics of intestinal stem cells using a novel
approach of continuous intravital imaging of Lgr5-Confetti mice. We find that Lgr5+ cells in the
upper part of the niche (termed ‘border cells’) can be passively displaced into the transit-
amplifying (TA) domain, following division of proximate cells, implying that determination of
stem cell fate can be uncoupled from division. Through the quantitative analysis of individual
clonal lineages, we show that stem cells at the crypt base, termed ‘central cells’, experience a
survival advantage over border stem cells. However, through the transfer of stem cells between the
border and central regions, all Lgr5+ cells are endowed with long-term self-renewal potential.
These findings establish a novel paradigm for stem cell maintenance in which a dynamically
heterogeneous cell population is able to function long-term as a single stem cell pool.
Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research,
subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms
Correspondence and requests for materials should be addressed to J.v.R. (j.vanrheenen@hubrecht.eu) or H.C.
(h.clevers@hubrecht.eu).
Author contributions J.v.R. and L.R. conceived the study. L.R. optimized the surgical and imaging procedure. L.R., S.I.J.E, A.Z, and
H.J.S. performed imaging experiments. L.R., H.J.S., B.D.S., S.I.J.E. performed analyses. F.R.S. provided the Lgr5DTR:EGFP mice
and B.D.S. did all biophysical modelling. L.R. and S.I.J.E. made the figures. J.v.R. and H.C. have supervised the study. All authors
discussed results and participated in preparation of the manuscript.
Full Methods and any associated references are available in the online version of the paper.
Reprints and permissions information is available at ww.nature.com/reprints.
The authors declare no competing financial interests Readers are welcome to comment on the online version of the paper.
Europe PMC Funders Group
Author Manuscript
Nature. Author manuscript; available in PMC 2014 September 20.
Published in final edited form as:
Nature. 2014 March 20; 507(7492): 362–365. doi:10.1038/nature12972.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
In the small intestine, stem cells are associated with Lgr5 expression, which marks around
14-16 proliferative ‘Crypt Base Columnar (CBC)’ cells distributed throughout the crypt
base. The stem cell niche is constituted by Paneth cells10,11 and surrounding mesenchyme12.
Cells that become displaced from this region enter the TA compartment and lose stemness13.
Quiescent or slow-cycling cells, positioned at or near the ‘+4 position’ may constitute a
second stem cell type3,5,6,14, although a recent study indicated that some, if not all, of these
cells represent secretory precursors that, in common with Dll1+ cells higher in the crypt15,
can be recruited back into the stem cell compartment upon damage16. Hierarchy,
heterogeneity, and spatial organization of intestinal stem cells remain a subject of
debate17-21. Are stem and progenitors organized in an engrained proliferative hierarchy,
defined by a signature of molecular markers, or do stem cells transit reversibly between
states of variable competence in which they become biased towards renewal or
differentiation? If the latter is true, is bias controlled by intrinsic heterogeneity in the
expression of fate determinants, or the consequence of spatio-temporal cues associated with
niche-derived signals? Although inducible genetic lineage tracing allows to dissect short-
term heterogeneity in self-renewal potential, its reliability may be undermined by transient
effects due to drug-inducing agents, Cre activity, or non-representativeness of labelling22.
Therefore we applied an in vivo live-imaging strategy, allowing measurements to begin
several days after drug administration. In common with previous live-imaging approaches
used to study stem cells in hair follicle and testis 23,24,25, our approach enables tracing of the
fate of individual marked stem cells and their progeny over time in vivo.
Multiphoton intravital microscopy and surgical implantation of an abdominal imaging
window (AIW)26,27 into living Lgr5EGFP-Ires-CreERT2/R26R-Confetti mice were used to
obtain visual access to the intestinal stem cell niche (Fig. 1a). Lgr5+ CBC cells and their
progeny were lineage traced over time (Extended Data Fig. 1) by activating the expression
of one of the Confetti colours (membranous CFP, cytoplasmic YFP and RFP) in individual
Lgr5+ cells using Tamoxifen-mediated recombination of the Confetti-construct (Fig. 1a). To
characterize fate behaviour of CBC cells we followed lineages of 80 marked cells (n = 4
mice) up to 5 days from the start of time-lapse imaging (Extended Data Fig. 2, for controls
see 27 and Extended Data Fig. 3).
Following induction, clonal progeny were observed throughout the stem cell niche. To
quantify fate behaviour of Lgr5+ CBC cells, we acquired Z-stacks (Fig. 1b; see Video 1 for
the 3D reconstruction) and classified cells based upon their relative position, using the most
basal cells (termed ‘row 0’) as a reference (Fig. 1b). Confetti-labelled clones were scored
according to cell number, disaggregated by position (Extended Data Fig. 4). In line with
predictions of neutral competition7, numbers of marked cells in the stem cell niche varied
widely between clones (some expanded in size, others lost attachment to this compartment
altogether; Extended Data Figs. 2 and 4). As just 1 of the 28 clones containing a single
marked CBC cell at the start of filming remained single after two days of tracing, we chose
to neglect the potential impact of lineage committed quiescent Lgr5+ cells, identified
previously16.
To investigate spatial heterogeneity in self-renewal potential of CBC cells, we defined two
regions within the Lgr5+ stem cell niche: a central (rows 0 to +2) and border (+3 and +4)
region (Fig. 1b). A ‘mother’ cell in either central or border region could expand and give
rise to progeny that extended into both regions (Fig. 1c-f and Extended Data Fig. 5). Further
quantitative analysis was necessary to address the potency of CBC cells in these two
domains. While the average number of central cells per clone derived from a single central
‘mother’ cell remained approximately constant, consistent with their maintenance over time,
the average number of border cells derived from these cells increased to approximately 2 by
day 3 (Fig. 2a). Furthermore, maintenance of the average central cell number was achieved
Ritsma et al. Page 2
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
through the steady decline in the number of clones retaining at least one central cell (Fig.
2b), compensated by a steady increase in size of those that remain (Fig. 2c). Although clones
derived from single border ‘mother’ cells also appeared to approximately maintain their
number, they gave rise to a comparatively smaller number of central cells (Fig. 2a). The
sustained increase in the number of border cells from a central ‘mother’ cell (Fig. 2a)
indicates that these cells typically outcompete cells at the niche border (Fig. 2d).
To investigate the potential basis of this positional advantage, we studied the development
of clones with finer time resolution. Every two hours, we acquired multiphoton images of
crypts, followed the location of all GFP-labelled cells over time (Extended Data Fig. 6;
Video 2-4) and found that division of single Lgr5+ cells coincides with displacement of
proximate CBC cells. This suggests that cell proliferation creates competition for space
leading to an adjustment of cell positions. Through this rearrangement, and independent of
their division history, CBC cells located at the border can become passively displaced from
the niche following division of a neighbour (Fig. 2e and Extended Data Fig. 4).
To challenge this conclusion and address the potency of the Lgr5+ CBC stem cell
population, we aimed to quantitatively capture the variability seen in the lineage potential of
individual cells by a biophysical model, involving a revision of the neutral drift dynamics
model introduced in [7,8] in which all stem cells were considered functionally equivalent. In
this new model, a periodic quasi-one-dimensional arrangement of stem cells mimicked the
‘collar-like’ geometry of the central and border niche regions of the crypt (Fig. 3a). To
account for the mixed GFP expression profile seen at rows +3 and +4 (Fig. 1b), the border
region was further subdivided into Lgr5+ CBC cells and Lgr5 TA cells. To accommodate
the range of observed dynamical behaviours in the stem cell niche, we allowed for five
possible ‘channels’ of stem cell loss and replacement (Fig. 3a): Following division of a
border stem cell, one daughter cell remains at its position while the other either (1),
displaces a border TA cell out of the niche; (2), displaces a border stem cell which in turn
displaces a border TA cell out of the niche; or (3), displaces a central stem cell which in turn
displaces a border stem cell into the border TA cell domain. Similarly, after division of a
central stem cell, one daughter remains at its position while the other either (4), displaces a
border stem cell into the border TA cell region; or (5), displaces a central cell which in turn
displaces a border stem cell into the TA cell region. If we define as λ the rate of transfer of
border TA cells out of the niche, each of these 5 processes occur at rates Pb
λ
, Pbb
λ
, Pbc
λ
,
Pcb
λ
, and Pcc
λ
, respectively, with Pb+Pbb+Pbc+Pcb+Pcc=1 (Supplementary Notes).
By fixing the relative rates of stem cell division and displacement by the observed average
clone size dependences and independent estimates of the average cell division rate, we
found that the biophysical model can accurately predict clone size distribution and spatial
dependencies observed in live-imaging (Fig. 2a-c and Fig. 3b, and Extended Data Fig. 7).
More significantly, with the same parameters, the model describes quantitatively
convergence onto the hallmark scaling behaviour reported using static lineage tracing assays
at intermediate times7 (7 and 14 days post-induction), as well as the predicted progression
towards crypt monoclonality at long-times8 (Extended Data Fig. 8 and 9; Supplementary
Notes).
To further challenge the model, we traced the recovery of stem cells following targeted
ablation of Lgr5+ cells using diphtheria toxin (DT) injection in mice where the human DT
receptor (DTR) fused to EGFP was knocked in the Lgr5 locus (Lgr5DTR:EGFP)28 (Fig. 4a).
In these mice, recovered Lgr5+ cells are derived from a TA-lineage28. Following complete
depletion (Fig. 4a), we observed a low frequency of initiation and a heterogeneous pattern of
recovery (Fig. 4b and Supplementary Video 5), suggesting sporadic transfer of cells from
the TA zone into the stem cell niche border. The cohesion of these recovered cell clusters
Ritsma et al. Page 3
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
(Supplementary Video 6) suggests clonal expansion of individual TA cells. Intriguingly, by
allowing individual border stem cells to recolonize a depleted stem cell niche through cell
division uncompensated by loss, our biophysical model provided a quantitative prediction of
cluster composition (border versus central) by size, with the same relative rates of stem cell
division as those found in steady-state (Fig. 4c and Supplementary Notes).
Our data shows that intestinal stem cell maintenance follows from competition between
proximate CBC stem cells for limited niche access and stem cells positioned near the niche
boundary experience a bias towards loss and replacement, while stem cells remote from the
boundary are biased towards survival. Intriguingly, a similar dependence of self-renewal
potential on proximity to the niche border was reported in a recent in vivo live-imaging
study of mouse hair follicle29, suggesting that such heterogeneity may be a ubiquitous
feature of adult stem cell populations. A recent lineage tracing study based on the
continuous and sporadic acquisition of mutations during DNA replication, concluded that
only a subfraction of putative intestinal stem cells are ‘functional’30. Our quantitative
analysis of live-imaging data shows that central stem cells are about 3 times more likely than
border cells to fully colonize a crypt in steady-state, explaining why only a fraction of Lgr5+
cells appears to retain long-term self-renewal potential (Supplementary Notes). Through the
transfer of cells between the central and border regions of the niche, the dynamic and
heterogeneous population of intestinal stem cells is able to function long-term as a single
equipotent pool.
Online-only Methods
Mice
All experiments were carried out in accordance with the guidelines of the Animal Welfare
Committee of the Royal Netherlands Academy of Arts and Sciences, the Netherlands. To
obtain R26R-Confetti; Lgr5-EGFP-Ires-CreERT2 mice, R26R-Confetti7 mice were crossed
with Lgr5-EGFP-Ires-CreERT21. Random double heterozygous male mice between 10-22
weeks old were used for experiments. Three days before imaging, mice were injected with
2.5-5 mg Tamoxifen (single injection; Sigma Aldrich) to induce activation of Cre
recombinase to induce expression of one of the Confetti colours (membranous CFP,
cytoplasmic YFP and RFP). Nuclear GFP was also activated, but that subset of confetti-
labelled cells was not followed. For the targeted ablation studies, 4 male Lgr5DTR:EGFP mice
carrying an AIW received 50 μg kg−1 diphtheria toxin (DT) through intraperitoneal
injections. Depletion of Lgr5+ cells was confirmed by intravital imaging. Mice in which
Lgr5+ cells were not completely depleted after 24 hours received a second DT injection.
Mice were housed under standard laboratory conditions and received food and water ad
libitum.
AIW surgery
The AIW surgery was performed as described in reference26. In short, all surgical
procedures were performed under 2% isoflurane (v/v) inhalation anaesthesia. Before
surgery, buprenorphine (3 μg per mouse; Temgesic©, BD pharmaceutical limited) was
administered intramuscularly. The left lateral flank of the mice was shaved and the skin was
disinfected with 70% (v/v) ethanol. Next, a left lateral flank incision was made through skin
and abdominal wall and a purse string suture was placed along the wound edge. A
disinfected AIW (> 1 hour in 70% (v/v) ethanol) was placed glass side down next to the
mice and the ileum was placed on top. 3M™ Vetbond™ Tissue Adhesive (n-butyl
cyanoacrylate; 3M) was used to fix the ileum to the cover glass of the AIW and CyGel
(BioStatus Limited) was added to diminish peristaltic movement. After 5 min the AIW was
inverted and placed in the mouse, with the skin and abdominal wall placed inside the AIW
Ritsma et al. Page 4
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
groove. Then sutures were tightened to stably secure the window into the animal. After
surgery the mice were provided food and water ad libitum. Furthermore, mice were closely
monitored once a day before imaging for behaviour, reactivity, appearance and defecation.
Equipment and settings
Intravital imaging was performed on an inverted Leica TCS SP5 AOBS two-photon
microscope with a chameleon Ti:Sapphire pumped optical parametric oscillator (Coherent
Inc.) equipped with a 25x (HCX IRAPO NA0.95 WD 2.5mm) water objective and four non-
descanned detectors (NDDs). The NDDs collect the following wavelengths: NDD1 <455
nm, NDD2 455-490 nm, NDD3 500-550 nm, NDD4 560-650 nm. Sequential scanning was
performed, exiting the tissue with 860 and 960 nm wavelengths. The Confetti colours were
detected as follows: 860 nm: NDD2 (CFP and eGFP), 960 nm: NDD3 (eGFP and YFP),
NDD4 (YFP and RFP). Second harmonic generation (SHG) signal is generated by 960 nm
excitation at collagen I and detected in NDD2. Scanning was performed in a bidirectional
mode at 700 Hz and 12 bit, with a zoom of 1.7, 512×512 pixels. Z-stacks with 2.5 μm z-
steps of typically 70-80 images were acquired. Re-identification of the same crypts over
multiple days was accomplished by storing the xy coordinates of the imaged regions using
the ‘multiple position’ function in the LAS-AF software and using the vasculature and the
typical (Confetti) Lgr5+ crypt pattern as visual landmarks.
Multi-day intestinal stem cell imaging
After placing the AIW the mice were kept under anaesthesia and placed face-down in a
custom-designed imaging box in which isoflurane (1% (v/v) was administered through a
facemask as described before26. For the multi-day imaging sessions (all imaging figures
except Extended Data Figure 6), mice were imaged once a day for a maximum of 3 hours
during which the climate chamber surrounding the microscope was kept at 32°C. After the
imaging session the mice were allowed to wake up to maintain their body temperature. After
imaging, acquired z-stacks were corrected for z and xy shifts using a custom-designed Visual
Basic software program and further processed and analysed using basic functions in ImageJ
software (linear contrasting, blurring, median filtering).
Short-term intestinal stem cell imaging
Mice were anesthetized using isoflurane (2% v/v). The left lateral flank was shaved and the
skin was disinfected using 70% (v/v) ethanol. Next, a left lateral flank incision was made
through skin and abdominal wall and the ileum was extracorporated using in PBS-drowned
cotton swabs. The ileum was placed on a custom-designed inset containing a coverslip
fitting the custom-designed imaging box. The ileum was secured to the cover slip using
Vetbond and CyGel. The mouse was placed on top of the intestine and in PBS-drowned
sterile cotton gauzes were placed next to the animal to prevent dehydration. Parafilm®M
(Sigma-Aldrich) was used to cover the mouse and a subcutaneous infusion system was used
to provide 100 μl of sterile PBS per hour. The inset was placed within the custom-designed
imaging box in which isoflurane (1%) v/v) was administered through a facemask as
described above. The temperature of the mouse was monitored during imaging using a rectal
probe and was kept between 36 and 37°C by adjusting the temperature of the surrounding
climate chamber. Imaging was performed every 2 hours for 14 hours. Z-stacks with a z-step
of 2.5 μm of 12 regions with on average 6 crypts were made. Acquired z-stacks were
analysed using ImageJ plugins (TurboReg, 3D visualization, 3D viewer).
Intravital imaging of Lgr5+ depleted mice
Mice carrying an AIW received 50 μg kg−1 diphtheria toxin (DT) through intraperitoneal
injections. Depletion of Lgr5+ cells was confirmed by intravital imaging. Mice in which
Ritsma et al. Page 5
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Lgr5+ cells were not completely depleted after 24 hours received a second DT injection.
Only mice in which full depletion was confirmed by intravital imaging 24 hours after the
last DT injection were analyzed. The number of Lgr5+-GFP cells within the stem cell niche
border and centre was determined as described in Fig. 1b.
Real-time lineage tracing – clonal competition strength and positional effect on clone size
The data from the lineage tracing was collected at random, and all clones that were imaged
with a 3-day interval were included. The strength of a Confetti-labelled Lgr5+ CBC cell to
produce offspring was expressed as the fold increase in Confetti-labelled Lgr5+ CBC cell
number three days after the first imaging session. A Mann Whitney U test was performed
because the data was not normally distributed.
Quantitative data analysis - Multi-day lineage tracing of Lgr5+ CBC cells
Lineage tracing was performed for 80 clones in 80 crypts from 4 mice. No sample size
estimate was calculated before the study was executed. Only data from mice from which
high enough quality images were acquired were included in the study. The number of
Confetti-labelled cells per crypt position (centre (rows 0 to +2), border (rows +3 to +4), TA
(rows >4)) was scored. From the 80 lineages, we obtained 33 sublineages originating from
the central region (Fig. 4A), and 47 sublineages from the border (Fig 4.B).
Immune cell analysis on intestinal tissue to test potential side-effects of AIW
Six E-Cadherin-CFP/Lgr5EGFP-Ires-CreERT2 mice, 22 weeks of age, were randomly divided
into two groups: a control and a window group. AIWs were implanted on top of the small
intestines of mice from the window group whereas mice from the control group did not
undergo surgery. After 24 hours all mice were sacrificed and the small intestines were
harvested. Note that in the window group the part of the small intestine that was located
directly behind the window was harvested. The small intestines were fixed for 1 day in
fixation mix (1% paraformaldehyde, 0.2% NaIO4, 61mM Na2HPO4, 75mM L-Lysine and
14 mM NaH2PO4 in H2O). After fixation, the tissues were placed for 6 hours into 30%
sucrose after which the tissues were snap-frozen using Tissue Freezing Medium (Leice
Microsystems Nussloch GmbH). 16 μm sections were cut using a Leica CM3050 cryotome.
A standard immunohistochemistry protocol was used to stain the sections with CD45
antibodies (BD Pharmingen™, 553078, Clone 30-F11) and random areas were imaged. For
analysis, 10 areas within the imaged regions were selected and analysed in a blinded
manner. The number of CD45 positive cells within a region was counted manually and an
averaged number for each mouse was calculated. Next, the average of the 3 mice per group
was calculated. A Mann-whitney U test was performed because the sample was not
distributed normally, and no significant differences were found. The variance between the
groups was tested with an F-test, and was not different.
Clone frequency window versus control mice to test potential side-effects of AIW
Eight Lgr5-EGFP-Ires-CreERT2 mice, 22 weeks of age, were divided into two groups: a
control and a window group. All mice received 5 mg Tamoxifen by intraperitoneal injection.
Three days later, AIWs were implanted on top of the small intestines of mice from the
window group whereas mice from the control group did not undergo surgery at this point.
Two days after the surgery (five days after Tamoxifen injection) all mice were imaged. In
the control group the intestine was exteriorized prior to imaging (as described in short-term
intestinal imaging). In the window group the mice were imaged through the AIW. Several
random areas were imaged. All recorded clones were used for analysis. For a single clone
the number of cells within the stem cell compartment was determined and a frequency
distribution was made for the two groups.
Ritsma et al. Page 6
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
The authors would like to thank Anko de Graaff from the Hubrecht Imaging Center for imaging support, all
members of the van Rheenen group for useful discussions and the Hubrecht Institute animal caretakers for animal
support. This work was supported by a Vidi fellowship (91710330; J.v.R.) and equipment grants (175.010.2007.00
and 834.11.002; J.v.R.) from the Dutch organization of scientific research (NWO), a grant from the Dutch cancer
society (KWF; HUBR 2009-4621; J.v.R.), a grant from the Association for International Cancer Research (AICR;
13-0297; J.v.R.), and the Wellcome Trust (grant number 098357/Z/12/Z; B.D.S.).
References
1. Barker N, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5.
Nature. 2007; 449:1003–1007. [PubMed: 17934449]
2. Sangiorgi E, Capecchi MR. Bmi1 is expressed in vivo in intestinal stem cells. Nat Genet. 2008;
40:915–920. [PubMed: 18536716]
3. Takeda N, et al. Interconversion Between Intestinal Stem Cell Populations in Distinct Niches.
Science. 2011; 334:1420–1424. [PubMed: 22075725]
4. Montgomery RK, et al. Mouse telomerase reverse transcriptase (mTert) expression marks slowly
cycling intestinal stem cells. Proceedings of the National Academy of Sciences. 2011; 108:179–184.
5. Powell, Anne E., et al. The Pan-ErbB Negative Regulator Lrig1 Is an Intestinal Stem Cell Marker
that Functions as a Tumor Suppressor. Cell. 2012; 149:146–158. [PubMed: 22464327]
6. Wong VWY, et al. Lrig1 controls intestinal stem-cell homeostasis by negative regulation of ErbB
signalling. Nat Cell Biol. 2012; 14:401–408. [PubMed: 22388892]
7. Snippert HJ, et al. Intestinal Crypt Homeostasis Results from Neutral Competition between
Symmetrically Dividing Lgr5 Stem Cells. Cell. 2010; 143:134–144. [PubMed: 20887898]
8. Lopez-Garcia C, Klein AM, Simons BD, Winton DJ. Intestinal Stem Cell Replacement Follows a
Pattern of Neutral Drift. Science. 2010; 330:822–825. [PubMed: 20929733]
9. Snippert HJ, Clevers H. Tracking adult stem cells. EMBO Rep. 2011; 12:113–122. [PubMed:
21252944]
10. Sato T, et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011;
469:415–418. [PubMed: 21113151]
11. VanDussen KL, et al. Notch signaling modulates proliferation and differentiation of intestinal crypt
base columnar stem cells. Development. 2012; 139:488–497. [PubMed: 22190634]
12. Farin HF, Van Es JH, Clevers H. Redundant Sources of Wnt Regulate Intestinal Stem Cells and
Promote Formation of Paneth Cells. Gastroenterology. 2012; 143:1518–1529.e1517. [PubMed:
22922422]
13. van der Flier LG, Clevers H. Stem Cells, Self-Renewal, and Differentiation in the Intestinal
Epithelium. Annual Review of Physiology. 2009; 71:241–260.
14. Sangiorgi E, Capecchi MR. Bmi1 lineage tracing identifies a self-renewing pancreatic acinar cell
subpopulation capable of maintaining pancreatic organ homeostasis. Proceedings of the National
Academy of Sciences. 2009; 106:7101–7106.
15. van Es JH, et al. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat Cell
Biol. 2012; 14:1099–1104. [PubMed: 23000963]
16. Buczacki SJA, et al. Intestinal label-retaining cells are secretory precursors expressing Lgr5.
Nature. 2013; 495:65–69. [PubMed: 23446353]
17. Stine RR, Matunis EL. Stem cell competition: finding balance in the niche. Trends in Cell Biology.
18. Simons, Benjamin D.; Clevers, H. Strategies for Homeostatic Stem Cell Self-Renewal in Adult
Tissues. Cell. 2011; 145:851–862. [PubMed: 21663791]
19. Morrison SJ, Spradling AC. Stem Cells and Niches: Mechanisms That Promote Stem Cell
Maintenance throughout Life. Cell. 2008; 132:598–611. [PubMed: 18295578]
Ritsma et al. Page 7
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
20. Goulas S, Conder R, Knoblich Juergen A. The Par Complex and Integrins Direct Asymmetric Cell
Division in Adult Intestinal Stem Cells. Cell stem cell. 2012; 11:529–540. [PubMed: 23040479]
21. Sheng XR, Matunis E. Live imaging of the Drosophila spermatogonial stem cell niche reveals
novel mechanisms regulating germline stem cell output. Development. 2011; 138:3367–3376.
[PubMed: 21752931]
22. Zhu Y, Huang Y-F, Kek C, Bulavin Dmitry V. Apoptosis Differently Affects Lineage Tracing of
Lgr5 and Bmi1 Intestinal Stem Cell Populations. Cell stem cell. 2013; 12:298–303. [PubMed:
23415913]
23. Nakagawa T, Sharma M, Nabeshima Y.-i. Braun RE, Yoshida S. Functional Hierarchy and
Reversibility Within the Murine Spermatogenic Stem Cell Compartment. Science. 2010; 328:62–
67. [PubMed: 20299552]
24. Klein AM, Nakagawa T, Ichikawa R, Yoshida S, Simons BD. Mouse Germ Line Stem Cells
Undergo Rapid and Stochastic Turnover. Cell stem cell. 2010; 7:214–224. [PubMed: 20682447]
25. Rompolas P, et al. Live imaging of stem cell and progeny behaviour in physiological hair-follicle
regeneration. Nature. 2012; 487:496–499. [PubMed: 22763436]
26. Ritsma L, et al. Surgical implantation of an abdominal imaging window for intravital microscopy.
Nat. Protocols. 2013; 8:583–594.
27. Ritsma L, et al. Intravital Microscopy Through an Abdominal Imaging Window Reveals a Pre-
Micrometastasis Stage During Liver Metastasis. Science Translational Medicine. 2012;
4:158ra145.
28. Tian H, et al. A reserve stem cell population in small intestine renders Lgr5-positive cells
dispensable. Nature. 2011; 478:255–259. [PubMed: 21927002]
29. Rompolas P, Mesa KR, Greco V. Spatial organization within a niche as a determinant of stem-cell
fate. Nature. 2013; 502:513–518. [PubMed: 24097351]
30. Kozar S, et al. Continuous Clonal Labeling Reveals Small Numbers of Functional Stem Cells in
Intestinal Crypts and Adenomas. Cell Stem Cell. 2013
Ritsma et al. Page 8
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 1. Intravital lineage tracing of Lgr5+ cells
a, Cartoon showing a mouse carrying an abdominal imaging window (AIW) to visualize
intestinal Lgr5+ CBC cells and their Confetti progeny over multiple imaging sessions. b,
Lateral projection of a Z-stack and representative XY-images of a crypt at indicated Z-stack
positions. The stem cell niche (Z0-13) is defined by Lgr5-GFP fluorescence. The relative
position of CBC cells to the most basal cell (row 0) determines location in the central (row 0
to +2, which translates to Z0-6) or border region (row +3 to +4, which translates to Z7-13)
of the stem cell niche. Scale bar, 20 μm. c-f, Intravital lineage tracing of RFP-expressing
Lgr5+ CBC cells located at the centre (c,d) and border (e,f) region. Grey lines indicate
crypts, white lines indicate Confetti clones. (d,f) Graphs show time evolution of spatial
organization of Confetti clones starting 3 days post-induction. Clone size is divided in
central (light green) and border (dark green) CBC cells. Asterisk indicates clones in which
all progeny were lost. Scale bar, 20 μm.
Ritsma et al. Page 9
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 2. Central CBC cells experience a short-term positional advantage in self-renewal
potential
a-c, Clonal evolution of a Confetti cell located at the central or border region starting 3 days
post-induction. Graphs show: a, average clone size; b, fraction “surviving” clones that
contain at least one marked central (top) or border (lower) cell; and c, average size of
surviving clones (clones with at least one marked cell). Different colours indicate different
regions in the niche. Points show data and lines show fit to the biophysical model (see Fig
3). Error bars represent s.d. d, Fold increase in clone size over three days from a border or
central Confetti+ CBC cell. Note that central stem cells have a positional advantage over
border stem cells. Error bars represent s.e.m., P <0.001 obtained using a Mann Whitney U
test. e, Intravital images of the same crypt at indicated times. Note that the yellow cell is
truly expelled from the stem cell niche, since GFP-expression was absent in the TA cell
region (see charts at indicated time points). Scale bars, 20 μm.
Ritsma et al. Page 10
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 3. Biophysical model of intestinal stem cell dynamics
a, From the unfolded crypt caricature (left), we synthesize a quasi-one-dimensional
biophysical model of the niche region (right) consisting of two domains: border and centre.
To conserve cell number, cell rearrangements following stem cell division displace precisely
one cell from the border. To capture the range of lineage data, we include 5 channels of stem
cell loss/replacement (1-5) defined in the main text. b, Cumulative size distributions of
clones derived from a single cell in the centre (left) or border (right). Clone size is defined in
both cases by total number of constituent cells in centre and border. Error bars represent
s.e.m. Points represent predictions of the model using the same parameters as that inferred
from the average dependences (Supplementary Notes).
Ritsma et al. Page 11
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 4. Recovery of stem cell compartment following ablation of Lgr5+ cells challenges model
a, Targeted ablation of Lgr5+ cells in Lgr5DTR:EGFP mice was induced by injection of
diphtheria toxin. Shown are representative images pre- and post-ablation. Scale bars, 20 μm.
b, Recovery of Lgr5+ CBC cells was monitored only in mice where full depletion was
confirmed 24 hours after diphtheria toxin injection. Images taken at 72 hours after depletion
show representative crypts containing clonal clusters of different sizes (n = 108 crypts in 3
mice). Scale bars, 20 μm c, For all various clone sizes, measured spatial composition (border
versus centre) of Lgr5+ CBC cells in clusters (grey) were accurately predicted by the
biophysical model (black). Error bars represent s.d.
Ritsma et al. Page 12
Nature. Author manuscript; available in PMC 2014 September 20.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
... Despite these fluctuations, the population level strategy of selfrenewal is found in many stem-cell systems [3]. Examples include the mammalian germline, the intestine, and the epidermis [6,[8][9][10][11]. In some organs, large variations of the number of stem cells can occur, such as for spermatogenic stem cells in murine testes [7]. ...
... Snippert et al. [10] and Lopez-Garcia et al. [9] demonstrated that the intestinal crypt uses neutral competition, where the progeny of one stem cell eventually takes over the entire niche. Ritsma et al. [11] and Corominas-Murtra [15] studied the dependence of lineage survival on cell position within the niche. ...
Article
Full-text available
Self-renewing tissues require that a constant number of proliferating cells is maintained over time. This maintenance can be ensured at the single-cell level or the population level. Maintenance at the population level leads to fluctuations in the number of proliferating cells over time. Often, it is assumed that those fluctuations can be reduced by increasing the number of asymmetric divisions, i.e., divisions where only one of the daughter cells remains proliferative. Here, we study a model of cell proliferation that incorporates a stem-cell niche of fixed size, and explicitly model the cells inside and outside the niche. We find that in this model, fluctuations are minimized when the difference in growth rate between the niche and the rest of the tissue is maximized and all divisions are symmetric divisions, producing either two proliferating or two nonproliferating daughters. We show that this optimal state leaves visible signatures in clone size distributions and could thus be detected experimentally.
... These adaptations of HSC fates are thought to be orchestrated by cell-extrinsic and cell-intrinsic factors. Extrinsic factors include the cellular microenvironment (10) and secreted factors (11) and were recently reviewed in detail (12,13). We thus focus here on the role of cell-intrinsic factors in HSC fate decision-making ( Figure 1). ...
Article
Full-text available
Hematopoietic stem cells (HSCs) can self-renew and differentiate for the entire life of an organism to produce new blood cells when needed. This process is regulated by asymmetric cell division (ACD), an evolutionarily conserved mechanism whereby cell fate determinants are unequally segregated into the daughter cells during division to instruct different cell fates. After many years of controversy, recent technical advances in microscopy, imaging, and bioinformatics make it now possible to visualize and quantify how factors segregate asymmetrically in dividing HSCs and lead to predictable changes in daughter cell fates many days later. While the molecular processes behind ACD in HSCs are still poorly understood, accumulating evidence suggests that lysosomes and other organelles, including mitochondria, autophagosomes, mitophagosomes, and recycling endosomes can segregate asymmetrically and act as cell fate determinants during divisions. Asymmetric segregation of lysosomes and mitochondria has been shown to predict mitochondrial activity, translation, and differentiation of HSC daughter cells and their offspring. This discovery and recent seminal findings show that lysosomes, once considered to be merely the trash bin of the cell, regulate many aspects of HSC biology and are crucial for the maintenance of quiescence and stem cell function. Here we provide a historical perspective and discuss the recent advances in our understanding of ACD and the role of lysosomes in HSC function. We discuss the limitations of past studies, talk about emerging concepts, and suggest critical next steps required to move the field forward.
... Previous studies based on mathematical modelling have shed light on quantitative mechanisms for homeostatic control via feedback when there is a one-way progression of differentiation from stem cells towards terminally differentiated cells (possibly via transient progenitors) (Johnston et al. 2007;Stiehl and Marciniak-Czochra 2011;Bocharov et al. 2011;Lander et al. 2009;Alarcon and Marciniak-Czochra 2011;Stiehl and Marciniak-Czochra 2017). However, it has been shown that differentiation is not always a one-way process, but priming or 'licensing' for differentiation may occur, which allows cells in early stages of differentiation to return to their naive stem cell state (Ritsma et al. 2014;Hara et al. 2014;Nakagawa et al. 2021;Greulich and Simons 2016;Chatzeli and Simons 2020). In this case, conditions for homeostasis may be more complex than previously proposed. ...
Article
Full-text available
To maintain renewing epithelial tissues in a healthy, homeostatic state, cell divisions and differentiation need to be tightly regulated. Mechanisms of homeostatic regulation often rely on crowding feedback control: cells are able to sense the cell density in their environment, via various molecular and mechanosensing pathways, and respond by adjusting division, differentiation, and cell state transitions appropriately. Here, we determine, via a mathematically rigorous framework, which general conditions for the crowding feedback regulation (i) must be minimally met, and (ii) are sufficient, to allow the maintenance of homeostasis in renewing tissues. We show that those conditions naturally allow for a degree of robustness toward disruption of regulation. Furthermore, intrinsic to this feedback regulation is that stem cell identity is established collectively by the cell population, not by individual cells, which implies the possibility of ‘quasi-dedifferentiation’, in which cells committed to differentiation may reacquire stem cell properties upon depletion of the stem cell pool. These findings can guide future experimental campaigns to identify specific crowding feedback mechanisms.
... Single-cell molecular profiling can be assessed through various techniques, including Fluorescence In Situ Hybridization (FISH) using fluorescence microscopy [122], quantitative Polymerase Chain Reaction (qPCR) with a qPCR reader [84], digital PCR (dPCR) employing a Biorad ddPCR droplet generator [123], DNA sequencing (DNA-seq) [124] and RNA sequencing (RNA-seq) using platforms like Illumina and Ion Torrent [125]. Additionally, proteomics and flow cytometry methods, carried out via flow cytometry, microfluidics, or CyTOF [126], as well as continuous live-cell imaging with fluorescence microscopy (epi or confocal) [127], offer valuable insights. The resolution of these techniques varies, with DNA and RNA sequencing providing genome-wide information, while dPCR and continuous live-cell imaging are suited for detecting fewer molecules (typically less than 10) within signaling pathways. ...
Article
Single-cell technology (SCT), which enables the examination of the fundamental units comprising biological organs, tissues, and cells, has emerged as a powerful tool, particularly in the field of biology, with a profound impact on stem cell research. This innovative technology opens new pathways for acquiring cell-specific data and gaining insights into the molecular pathways governing organ function and biology. SCT is not only frequently used to explore rare and diverse cell types, including stem cells, but it also unveils the intricacies of cellular diversity and dynamics. This perspective, crucial for advancing stem cell research, facilitates non-invasive analyses of molecular dynamics and cellular functions over time. Despite numerous investigations into potential stem cell therapies for genetic disorders, degenerative conditions, and severe injuries, the number of approved stem cell-based treatments remains limited. This limitation is attributed to the various heterogeneities present among stem cell sources, hindering their widespread clinical utilization. Furthermore, stem cell research is intimately connected with cutting-edge technologies, such as microfluidic organoids, CRISPR technology, and cell/tissue engineering. Each strategy developed to overcome the constraints of stem cell research has the potential to significantly impact advanced stem cell therapies. Drawing from the advantages and progress achieved through SCT-based approaches, this study aims to provide an overview of the advancements and concepts associated with the utilization of SCT in stem cell research and its related fields.
... In order to assess if Lgr5 +CBC cells in NMRs entered quiescence (G0), we used p27, a cyclin-dependent kinase inhibitor, that modulates the probability that cells will exit the active cell cycle after a mitotic division 40,41 . Firstly, we found no Lgr5 +CBC cells that were also positive for p27 in the mouse small intestine (duodenum), in agreement with the actively cycling status of mouse ASCs 31,35,42 (Fig. 2c). Similarly, we detected no p27 positivity in the NMR Lgr5 +CBC cells of the duodenum (Fig. 2c). ...
Article
Full-text available
The naked mole rat (NMR), Heterocephalus glaber, the longest-living rodent, provides a unique opportunity to explore how evolution has shaped adult stem cell (ASC) activity and tissue function with increasing lifespan. Using cumulative BrdU labelling and a quantitative imaging approach to track intestinal ASCs (Lgr5+) in their native in vivo state, we find an expanded pool of Lgr5+ cells in NMRs, and these cells specifically at the crypt base (Lgr5+CBC) exhibit slower division rates compared to those in short-lived mice but have a similar turnover as human LGR5+CBC cells. Instead of entering quiescence (G0), NMR Lgr5+CBC cells reduce their division rates by prolonging arrest in the G1 and/or G2 phases of the cell cycle. Moreover, we also observe a higher proportion of differentiated cells in NMRs that confer enhanced protection and function to the intestinal mucosa which is able to detect any chemical imbalance in the luminal environment efficiently, triggering a robust pro-apoptotic, anti-proliferative response within the stem/progenitor cell zone.
Article
Full-text available
Inflammatory bowel disease (IBD), including Crohn’s disease and ulcerative colitis, is a disease of chronic inflammatory conditions of the intestinal tract due to disturbance of the inflammation and immune system. Symptoms of IBD include abdominal pain, diarrhea, bleeding, reduced weight, and fatigue. In IBD, the immune system attacks the intestinal tract’s inner wall, causing chronic inflammation and tissue damage. In particular, interlukin-6 and interlukin-17 act on immune cells, including T cells and macrophages, to amplify the immune responses so that tissue damage and morphological changes occur. Of note, excessive calorie intake and obesity also affect the immune system due to inflammation caused by lipotoxicity and changes in lipids supply. Similarly, individuals with IBD have alterations in liver function after sustained high-fat diet feeding. In addition, excess dietary fat intake, along with alterations in primary and secondary bile acids in the colon, can affect the onset and progression of IBD because inflammatory cytokines contribute to insulin resistance; the factors include the release of inflammatory cytokines, oxidative stress, and changes in intestinal microflora, which may also contribute to disease progression. However, interfering with de novo fatty acid synthase by deleting the enzyme acetyl-CoA-carboxylase 1 in intestinal epithelial cells (IEC) leads to the deficiency of epithelial crypt structures and tissue regeneration, which seems to be due to Lgr5+ intestinal stem cell function. Thus, conflicting reports exist regarding high-fat diet effects on IBD animal models. This review will focus on the pathological basis of the link between dietary lipids intake and IBD and will cover the currently available pharmacological approaches.
Article
Full-text available
The complex functioning of multi-cellular tissue development relies on proper cell production rates to replace dead or differentiated specialized cells. Stem cells are critical for tissue development and maintenance, as they produce specialized cells to meet the tissues’ demands. In this study, we propose a computational model to investigate the stem cell’s mechanism, which generates the appropriate proportion of specialized cells, and distributes them to their correct position to form and maintain the organized structure in the population through intercellular reactions. Our computational model focuses on early development, where the populations overall behavior is determined by stem cells and signaling molecules. The model does not include complicated factors such as movement of specialized cells or outside signaling sources. The results indicate that in our model, the stem cells can organize the population into a desired spatial pattern, which demonstrates their ability to self-organize as long as the corresponding leading signal is present. We also investigate the impact of stochasticity, which provides desired non-genetic diversity; however, it can also break the proper boundaries of the desired spatial pattern. We further examine the role of the death rate in maintaining the system’s steady state. Overall, our study sheds light on the strategies employed by stem cells to organize specialized cells and maintain proper functionality. Our findings provide insight into the complex mechanisms involved in tissue development and maintenance, which could lead to new approaches in regenerative medicine and tissue engineering.
Article
All adult tissues experience wear and tear. Most tissues can compensate for cell loss through the activity of resident stem cells. Although the cellular maintenance strategies vary greatly between different adult (read: postnatal) tissues, the function of stem cells is best defined by their capacity to replace lost tissue through division. We discuss a set of six complementary hallmarks that are key enabling features of this basic function. These include longevity and self-renewal, multipotency, transplantability, plasticity, dependence on niche signals, and maintenance of genome integrity. We discuss these hallmarks in the context of some of the best-understood adult stem cell niches.
Article
The human oral mucosa contains one of the most complex cellular systems that are essential for normal physiology and defense against a wide variety of local pathogens. Evolving techniques and experimental systems have helped refine our understanding of this complex cellular network. Current single-cell RNA sequencing methods can resolve subtle differences between cell types and states, thus providing a great tool for studying the molecular and cellular repertoire of the oral mucosa in health and disease. However, it requires the dissociation of tissue samples, which means that the interrelationships between cells are lost. Spatial transcriptomic methods bypass tissue dissociation and retain this spatial information, thereby allowing gene expression to be assessed across thousands of cells within the context of tissue structural organization. Here, we discuss the contribution of spatial technologies in shaping our understanding of this complex system. We consider the impact on identifying disease cellular neighborhoods and how space defines cell state. We also discuss the limitations and future directions of spatial sequencing technologies with recent advances in machine learning. Finally, we offer a perspective on open questions about mucosal homeostasis that these technologies are well placed to address.
Article
Full-text available
Stem-cell niches in mammalian tissues are often heterogeneous and compartmentalized; however, whether distinct niche locations determine different stem-cell fates remains unclear. To test this hypothesis, here we use the mouse hair follicle niche and combine intravital microscopy with genetic lineage tracing to re-visit the same stem-cell lineages, from their exact place of origin, throughout regeneration in live mice. Using this method, we show directly that the position of a stem cell within the hair follicle niche can predict whether it is likely to remain uncommitted, generate precursors or commit to a differentiated fate. Furthermore, using laser ablation we demonstrate that hair follicle stem cells are dispensable for regeneration, and that epithelial cells, which do not normally participate in hair growth, re-populate the lost stem-cell compartment and sustain hair regeneration. This study provides a general model for niche-induced fate determination in adult tissues.
Article
Full-text available
The rapid cell turnover of the intestinal epithelium is achieved from small numbers of stem cells located in the base of glandular crypts. These stem cells have been variously described as rapidly cycling or quiescent. A functional arrangement of stem cells that reconciles both of these behaviours has so far been difficult to obtain. Alternative explanations for quiescent cells have been that they act as a parallel or reserve population that replace rapidly cycling stem cells periodically or after injury; their exact nature remains unknown. Here we show mouse intestinal quiescent cells to be precursors that are committed to mature into differentiated secretory cells of the Paneth and enteroendocrine lineage. However, crucially we find that after intestinal injury they are capable of extensive proliferation and can give rise to clones comprising the main epithelial cell types. Thus, quiescent cells can be recalled to the stem-cell state. These findings establish quiescent cells as an effective clonogenic reserve and provide a motivation for investigating their role in pathologies such as colorectal cancers and intestinal inflammation.
Article
Full-text available
Lgr5(+) intestinal stem cells generate enterocytes and secretory cells. Secretory lineage commitment requires Notch silencing. The Notch ligand Dll1 is expressed by a subset of immediate stem cell daughters. Lineage tracing in Dll1(GFP-ires-CreERT2) knock-in mice reveals that single Dll1(high) cells generate small, short-lived clones containing all four secretory cell types. Lineage specification thus occurs in immediate stem cell daughters through Notch lateral inhibition. Cultured Dll1(high) cells form long-lived organoids (mini-guts) on brief Wnt3A exposure. When Dll1(high) cells are genetically marked before tissue damage, stem cell tracing events occur. Thus, secretory progenitors exhibit plasticity by regaining stemness on damage.
Article
Full-text available
The adult Drosophila midgut is maintained by intestinal stem cells (ISCs) that generate both self-renewing and differentiating daughter cells. How this asymmetry is generated is currently unclear. Here, we demonstrate that asymmetric ISC division is established by a unique combination of extracellular and intracellular polarity mechanisms. We show that Integrin-dependent adhesion to the basement membrane induces cell-intrinsic polarity and results in the asymmetric segregation of the Par proteins Par-3, Par-6, and aPKC into the apical daughter cell. Cell-specific knockdown and overexpression experiments suggest that increased activity of aPKC enhances Delta/Notch signaling in one of the two daughter cells to induce terminal differentiation. Perturbing this mechanism or altering the orientation of ISC division results in the formation of intestinal tumors. Our data indicate that mechanisms for intrinsically asymmetric cell division can be adapted to allow for the flexibility in lineage decisions that is required in adult stem cells.
Article
Lineage-tracing approaches, widely used to characterize stem cell populations, rely on the specificity and stability of individual markers for accurate results. We present a method in which genetic labeling in the intestinal epithelium is acquired as a mutation-induced clonal mark during DNA replication. By determining the rate of mutation in vivo and combining this data with the known neutral-drift dynamics that describe intestinal stem cell replacement, we quantify the number of functional stem cells in crypts and adenomas. Contrary to previous reports, we find that significantly lower numbers of "working" stem cells are present in the intestinal epithelium (five to seven per crypt) and in adenomas (nine per gland), and that those stem cells are also replaced at a significantly lower rate. These findings suggest that the bulk of tumor stem cell divisions serve only to replace stem cell loss, with rare clonal victors driving gland repopulation and tumor growth.
Article
Adult stem cells reside in local microenvironments (niches) that produce signals regulating the outcome of stem cell divisions and stem cell-niche interactions. Limited space and signals in the niche often force stem cells to compete with one another. Although previous studies have uncovered several examples of genetically distinct stem cells competing for niche access, recent studies demonstrate that genetically equivalent stem cells compete under normal conditions, resulting in dynamic stem cell behavior in the niche. New work in multiple vertebrate and invertebrate tissues shows that stem cell competition occurs continuously and mutations disrupting the balance between competing stem cells can cause diseases and defects in the niche. This review discusses recent insights into stem cell competition in mammals and Drosophila.
Article
High-resolution intravital microscopy through imaging windows has become an indispensable technique for the long-term visualization of dynamic processes in living animals. Easily accessible sites such as the skin, the breast and the skull can be imaged using various different imaging windows; however, long-term imaging studies on cellular processes in abdominal organs are more challenging. These processes include colonization of the liver by metastatic tumor cells and the development of an immune response in the spleen. We have recently developed an abdominal imaging window (AIW) that allows long-term imaging of the liver, the pancreas, the intestine, the kidney and the spleen. Here we describe the detailed protocol for the optimal surgical implantation of the AIW, which takes ∼1 h, and subsequent multiphoton imaging, which takes up to 1 month.
Article
Emerging lineage-tracing data support the existence of several pools of intestinal stem cells (ISCs) in the adult mouse. The +4 location is known to harbor proliferative cells undergoing robust apoptosis in response to irradiation, but their relationship with recently reported ISC models is unclear. Here, we found that tamoxifen, at doses commonly used to induce lineage tracing, mimics the irradiation-induced apoptotic response of the +4 cells. We found that about 40% of apoptotic cells were Lgr5-positive whereas Bmi1-positive ISCs became sensitive to tamoxifen upon entering a proliferative state. In turn, when we suppressed apoptosis by either Bcl2 overexpression or Chk2 deletion, we found that lineage tracing of Lgr5-positive cells was efficiently reduced. In contrast, lineage tracing from Bmi1-positive ISCs was substantially increased in apoptosis-deficient backgrounds. We propose that apoptosis plays an important role in controlling lineage tracing from different ISC populations in the mouse intestine.
Article
Cell dynamics in subcutaneous and breast tumors can be studied through conventional imaging windows with intravital microscopy. By contrast, visualization of the formation of metastasis has been hampered by the lack of long-term imaging windows for metastasis-prone organs, such as the liver. We developed an abdominal imaging window (AIW) to visualize distinct biological processes in the spleen, kidney, small intestine, pancreas, and liver. The AIW can be used to visualize processes for up to 1 month, as we demonstrate with islet cell transplantation. Furthermore, we have used the AIW to image the single steps of metastasis formation in the liver over the course of 14 days. We observed that single extravasated tumor cells proliferated to form "pre-micrometastases," in which cells lacked contact with neighboring tumor cells and were active and motile within the confined region of the growing clone. The clones then condensed into micrometastases where cell migration was strongly diminished but proliferation continued. Moreover, the metastatic load was reduced by suppressing tumor cell migration in the pre-micrometastases. We suggest that tumor cell migration within pre-micrometastases is a contributing step that can be targeted therapeutically during liver metastasis formation.
Article
Background & aims: Wnt signaling regulates multiple aspects of intestinal physiology, including stem cell maintenance. Paneth cells support stem cells by secreting Wnt, but little is known about the exact sources and primary functions of individual Wnt family members. Methods: We analyzed intestinal tissues and cultured epithelial cells from adult mice with conditional deletion of Wnt3 (Vil-CreERT2;Wnt3fl/fl mice). We also analyzed intestinal tissues and cells from Atoh1 mutant mice, which lack secretory cells. Results: Unexpectedly, Wnt3 was dispensable for maintenance of intestinal stem cells in mice, indicating a redundancy of Wnt signals. By contrast, cultured crypt organoids required Paneth cell-derived Wnt3. Addition of exogenous Wnt, or coculture with mesenchymal cells, restored growth of Vil-CreERT2;Wnt3fl/fl crypt organoids. Intestinal organoids from Atoh1 mutant mice did not grow or form Paneth cells; addition of Wnt3 allowed growth in the absence of Paneth cells. Wnt signaling had a synergistic effect with the Lgr4/5 ligand R-spondin to induce formation of Paneth cells. Mosaic expression of Wnt3 in organoids using a retroviral vector promoted differentiation of Paneth cells in a cell-autonomous manner. Conclusions: Wnt is part of a signaling loop that affects homeostasis of intestinal stem and Paneth cells in mice. Wnt3 signaling is required for growth and development of organoid cultures, whereas nonepithelial Wnt signals could provide a secondary physiological source of Wnt.