Supplementary Material

Hierarchy and Plasticity in the Intestinal Stem Cell Compartment

Maryam Yousefi1, Linheng Li2 and Christopher J. Lengner1

1Department of Biomedical Sciences, School of Veterinary Medicine and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104

2Stowers Institute for Medical Research, Kansas City, Missouri, 64110, USA

Corresponding author: Lengner, C.J. (); Li, L. ()

Table S1. Proxy Reporter Alleles Marking Intestinal Stem Cell Populations, Their Specificity, and Contribution toIntestinal Regeneration and Oncogenesis.

Marker / Reporter Type / Marked Cell Position / Marked Cell Function (Assay) / Regeneration under basal conditions / Function during regeneration (Injury model) / Tumor initiation capacity (model) / Ref
Lgr5
Lgr5-eGFP-IRES-CreER (CreER) / Targeted knock-in / Crypt base / CBCs (Lineage tracing) / Frequent lineage tracing / Gene ablation in Lgr5 ISCs impairs regeneration (12 Gy gamma IR), lineage tracing after etiposide treatment / hyperplasia uponAPCflox/flox deletion / [1–5]
Lgr5-eGFP-IRES-CreER (eGFP) / Targeted knock-in / Crypt base and above / CBCs &T/A cells (Organoid formation) / N/A / Lgr5-eGFPHigh cells are radiosensitive (12 Gy gamma IR) / N/A / [1,2,6,7]
Lgr5-LacZ / Targeted knock-in / Crypt base and above / Undetermined / N/A / N/A / N/A / [1]
Lgr5-dsRED-IRES-CreERT2 / Targeted knock-in / Crypt base and above / CBCs (Lineage tracing) / Lineage tracing up to 60 days after CreER induction / N/A / N/A / [8]
Lgr5-DTR-eGFP / Targeted knock-in / Crypt base and above / CBCs (Organoid formation) / Cell ablation is tolerated in vivo and in vitro / Cell ablation impairs regeneration after 10 Gy radiation, but not following DSS / APC loss in the epithelium drives hyperplasia in the absence of Lgr5+ cells / [8,9]
mRNA / Endogenous / Crypt base and above / Undetermined / N/A / N/A / N/A / [8,10,11]
Protein / Endogenous / Crypt base (Human) / Undetermined / N/A / N/A / N/A / [12]
Bmi1
Bmi1-CreER / Targeted knock-in / Crypt cells from the +2 to +8 position and rare cells within villi / Reserve ISC (Lineage Tracing/Organoid formation/Cell ablation) / Lineage tracing up to 12 months, Bmi1-Cre+ cell ablation results in crypt loss / Contributes to regeneration following irradiation (12 Gy gamma IR) and etiposide treatment / Adenomas upon deletion of Ctnnb1exon3flox / [2,5,13]
Bmi1-CreER / Random integrant BAC transgenic / Single cells at position +3 to +6 / Reserve ISC (Lineage Tracing) / CBC generation under basal conditions / Increased CBC generation and lineage tracing after CBC cell ablation / N/A / [8]
Bmi1-GFP / Targeted knock-in / Position +3 to +6 (IF for GFP), undetectable by flow cytometry / Undetermined / N/A / Increase in frequency upon CBC ablation / N/A / [2,8]
mRNA / Endogenous / Crypt base and above / Undetermined / N/A / N/A / N/A / [2,11,14]
Protein / Endogenous / Around the +4 position / Undetermined / N/A / N/A / N/A / [15]
Hopx
Hopx-CreER / Targeted knock-in / Rare cells at or near the +4 position / Reserve ISC (Lineage Tracing/Organoid formation) / Lineage tracing with no clonal extinction 6 months after induction / Gene ablation in Hopx ISCs impairs regeneration (12 Gy gamma IR), lineage tracing after etiposide treatment / N/A / [2,5,16]
Hopx-eGFP / Targeted knock-in / Crypt base and above / Reserve ISCs, Active CBCs, Secretory cells (Organoid formation) / N/A / N/A / N/A / [2]
mRNA / Endogenous / Crypt base and above / Undetermined / N/A / N/A / N/A / [2,11]
Protein / Endogenous / Present in Lgr5-eGFP+ cells (proteomic analysis) / Undetermined / N/A / N/A / N/A / [10]
Lrig1
Lrig1-CreER / Targeted knock-in / Scattered single cells throughout crypt / Reserve and active ISCs (lineage tracing) / Lineage tracing up to 90 days, decreasing tracing events over time / Increased lineage tracing after 8 Gy x-ray injury / APC loss drives intestinal adenomas and colonal tumors / [17]
Lrig1-mApple / Random integrant BAC transgenic / Crypt base and above / Undetermined / N/A / N/A / N/A / [18]
mRNA / Endogenous / Crypt base and above (more Lrig1 mRNA in Lgr5 cells than Lrig-CreER cells) / Undetermined / N/A / N/A / N/A / [2,11,17,19]
Protein / Endogenous
Lrig1- antibody / Recognizes glycosylated & non-glycosylated Lrig1 / Crypt base / Undetermined / N/A / N/A / N/A / [10,18,19]
Lrig1-VU antibody / Recognizes non-glycosylated form of Lrig1 / One to three cells at the crypt base and occasionally cells higher up the crypt / Some overlap with Lgr5 CBCs (gene expression) / N/A / N/A / N/A / [17,18]
mTert
mTert–CreER / Random integrant transgenic / Single or small clusters of cells around +4 position / Reserve ISs (Lineage tracing) / Lineage tracing is detected even after one year / Tracing following 1, 10, and 15 Gy gamma-IR / N/A / [20]
mTert-eGFP / Random integrant transgenic / Rare cells at position +5 to +8, one marked cell per 150 crypts / Undetermined / N/A / Resistant to 10 Gy gamma-IR / N/A / [20,21]
mRNA / Endogenous / Crypt base / Undetermined / N/A / N/A / N/A / [2,10,14]
Protein / Endogenous / N/A / Undetermined / N/A / N/A / N/A
Sox9
Sox9-CreERT2 / Targeted knock-in / Crypt base / Reserve ISCs, CBCs, Paneth cells (lineage tracing) / Frequent lineage tracing / Tracing following 14 Gy x-ray radiation / N/A / [22]
Sox9-eGFP / Random integrant BAC transgenic / Crypt base / Reserve ISCs, CBCs, secretory progenitors (organoid formation, expression) / N/A / N/A / N/A / [22,23]
mRNA / Endogenous / Crypt base / N/A / N/A / N/A / N/A / [2,11,14]
Protein / Endogenous / Crypt base and some cells in the villi / N/A / N/A / N/A / N/A / [24,25]
Msi1
Msi1-eGFP / Random integrant transgenic / Crypt base and above / N/A / N/A / N/A / N/A / [26]
mRNA / Endogenous / Crypt base and above / N/A / N/A / N/A / N/A / [2,11,14]
Protein / Endogenous / Crypt base and above / N/A / N/A / Msi1 protein is upregulated during regeneration / N/A / [2,3,27,28]
DCLK1
DCKL1-CreER / Random integrant BAC transgenic / Rare cells around +4 position in the crypt / Tuft cells (Immunostaining), rarely long-lived (lineage tracing) / Rare lineage tracing. Cell ablation tolerated in vivo. / Cell ablation impairs regeneration (12 Gy radiation or 3% DSS; tracing is rare) / Colon tumor only after both deletion of APC and DSS treatment / [29]
DCKL1-CreER / Targeted knock-in / Scattered cells, mostly in the lower crypt / Tuft cells(Immunostaining) / No lineage tracing, cell ablation tolerated in vivo. / Rare tracing after radiation 8Gy or DSS (2%) / Polyp collapse after cell ablation, no adenoma in Ctnnb1exon3flox model / [30]
mRNA / Endogenous / Isolated cells from the base of the crypts up to villi / Tuft cells (Transcript counting) / N/A / N/A / N/A / [14]
Protein / Endogenous / Rare cells at crypt base, +4 position, and villi / Some CBCs and Tuft cells (Immunostaining for Tuft cell markers) / N/A / Protein not detected during regeneration following 12 Gy gamma IR / Limited expression pattern in APC min tumors / [29,31,32]
Axin2
Axin2-CreERT2-tdTomato(CreER) / Targeted knock-in / Crypt base and above / CBCs, Reserve ISCs, and TA cells / Frequent lineage tracing events / Frequent lineage tracing following 12 Gy gamma-IR / N/A / [33]
Axin2-CreERT2-tdTomato (Tomato) / Targeted knock-in / Crypt base and above / CBCs, Reserve ISCs, and TA cells / N/A / N/A / N/A / [34]
Axin2-LacZ / Targeted knock-in / Crypt base and above / Crypt cells except Paneth cells / N/A / N/A / N/A / [25,35]
mRNA / Endogenous / Crypt base / CBCS and Reserve ISCs (single cell gene expression analysis) / N/A / N/A / N/A / [34,36]
Protein / Endogenous / Crypt base / Undetermined / N/A / N/A / N/A / [36]
Dll1
Dll1-GFP-IRES-CreERT (CreER) / Targeted knock-in / Single cells around +5 position / Secretory progenitors (lineage tracing) / Rare lineage tracing (~10 per mouse) / Rare lineage tracing after injury (6 Gy gamma-IR) / N/A / [37]
Dll1-GFP-IRES-CreERT(GFP) / Targeted knock-in / Paneth cells and some cells higher in the crypt and villi / Paneth cells, secretory progenitors, and goblet cells (histology, expression) / Rarely generate Lgr5+ ISCs. Organoid formation only with Wnt3a / N/A / [37]
mRNA / Endogenous / Paneth cells and some cells higher in the crypt and villi / N/A / N/A / N/A / N/A / [37]
Protein / Endogenous / N/A / N/A / N/A / N/A / N/A
Alpi
Alpi-IRES-CreERT2 / Targeted knock-in / Cells in the villi / Enterocytes / No tracing under basal condition / Tracing after ablation of CBCs / No tumor formation after APC/Kras mutation / [38]
mRNA / Endogenous / Cells in the villi / Enterocytes / N/A / N/A / N/A / [38]
Protein / Endogenous / Cells in the villi / Enterocytes / N/A / N/A / N/A / [39]
Label retention
TetO-H2B-eGFP / Targeted knock-in (safe haven chromatin) / Crypt base and above at +3 to +5 positions / Paneth Cells, enteroendocrine cells, secretory progenitors, and rare reserve ISCs / N/A / No EdU incorporation post-12Gy gamma-IR / N/A / [11]
Cyp1a1-H2B-YFP / Random integrant transgenic / Crypt base and above at +3 to +5 positions / Paneth Cells, secretory progenitors / 2 day lineage tracing / Very rare lineage tracing (~10 events per mouse) after injury / N/A / [40]

N/A: Not applicable

Reference

1 Barker, N. et al. (2007) Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007

2 Li, N. et al. (2014) Single-cell analysis of proxy reporter allele-marked epithelial cells establishes intestinal stem cell hierarchy. Stem Cell Rep. 3, 876–891

3 Yousefi, M. et al. (2016) Msi RNA-binding proteins control reserve intestinal stem cell quiescence. J. Cell Biol. 215, 401–413

4 Barker, N. et al. (2009) Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457, 608–611

5 Tinkum, K.L. et al. (2015) Fasting protects mice from lethal DNA damage by promoting small intestinal epithelial stem cell survival. Proc. Natl. Acad. Sci. U. S. A. 112, E7148-7154

6 Merlos-Suárez, A. et al. (2011) The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell 8, 511–524

7 Tao, S. et al. (2015) Wnt activity and basal niche position sensitize intestinal stem and progenitor cells to DNAdamage. EMBO J. 34, 624–640

8 Tian, H. et al. (2011) A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478, 255–259

9 Metcalfe, C. et al. (2014) Lgr5+ stem cells are indispensable for radiation-induced intestinal regeneration. Cell Stem Cell 14, 149–159

10 Muñoz, J. et al. (2012) The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent “+4” cell markers. EMBO J. 31, 3079–3091

11 Li, N. et al. (2016) Mouse Label-Retaining Cells Are Molecularly and Functionally Distinct From Reserve Intestinal Stem Cells. Gastroenterology 151, 298–310.e7

12 Junttila, M.R. et al. (2015) Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer. Sci. Transl. Med. 7, 314ra186

13 Sangiorgi, E. and Capecchi, M.R. (2008) Bmi1 is expressed in vivo in intestinal stem cells. Nat. Genet. 40, 915–920

14 Itzkovitz, S. et al. (2011) Single-molecule transcript counting of stem-cell markers in the mouse intestine. Nat. Cell Biol. 14, 106–114

15 Srinivasan, T. et al. (2016) Notch signalling regulates asymmetric division and inter-conversion between lgr5 and bmi1 expressing intestinal stem cells. Sci. Rep. 6, 26069

16 Takeda, N. et al. (2011) Interconversion between intestinal stem cell populations in distinct niches. Science 334, 1420–1424

17 Powell, A.E. et al. (2012) The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell 149, 146–158

18 Poulin, E.J. et al. (2014) Using a new Lrig1 reporter mouse to assess differences between two Lrig1 antibodies in the intestine. Stem Cell Res. 13, 422–430

19 Wong, V.W.Y. et al. (2012) Lrig1 controls intestinal stem-cell homeostasis by negative regulation of ErbB signalling. Nat. Cell Biol. 14, 401–408

20 Montgomery, R.K. et al. (2011) Mouse telomerase reverse transcriptase (mTert) expression marks slowly cycling intestinal stem cells. Proc. Natl. Acad. Sci. U. S. A. 108, 179–184

21 Breault, D.T. et al. (2008) Generation of mTert-GFP mice as a model to identify and study tissue progenitor cells. Proc. Natl. Acad. Sci. U. S. A. 105, 10420–10425

22 Roche, K.C. et al. (2015) SOX9 maintains reserve stem cells and preserves radioresistance in mouse small intestine. Gastroenterology 149, 1553–1563.e10

23 Ramalingam, S. et al. (2012) Distinct levels of Sox9 expression mark colon epithelial stem cells that form colonoids in culture. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G10-20

24 Gracz, A.D. et al. (2010) Sox9 expression marks a subset of CD24-expressing small intestine epithelial stem cells that form organoids in vitro. Am. J. Physiol. Gastrointest. Liver Physiol. 298, G590-600

25 Sato, T. et al. (2011) Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469, 415–418

26 Maria Cambuli, F. et al. (2013) Brief report: musashi1-eGFP mice, a new tool for differential isolation of the intestinal stem cell populations. Stem Cells Dayt. Ohio 31, 2273–2278

27 Potten, C.S. et al. (2003) Identification of a putative intestinal stem cell and early lineage marker; musashi-1. Differ. Res. Biol. Divers. 71, 28–41

28 Wang, S. et al. (2015) Transformation of the intestinal epithelium by the MSI2 RNA-binding protein. Nat. Commun. 6, 6517

29 Westphalen, C.B. et al. (2014) Long-lived intestinal tuft cells serve as colon cancer-initiating cells. J. Clin. Invest. 124, 1283–1295

30 Nakanishi, Y. et al. (2013) Dclk1 distinguishes between tumor and normal stem cells in the intestine. Nat. Genet. 45, 98–103

31 May, R. et al. (2008) Identification of a novel putative gastrointestinal stem cell and adenoma stem cell marker, doublecortin and CaM kinase-like-1, following radiation injury and in adenomatous polyposis coli/multiple intestinal neoplasia mice. Stem Cells Dayt. Ohio 26, 630–637

32 May, R. et al. (2009) Doublecortin and CaM kinase-like-1 and leucine-rich-repeat-containing G-protein-coupled receptor mark quiescent and cycling intestinal stem cells, respectively. Stem Cells Dayt. Ohio 27, 2571–2579

33 Li, N. et al. (2016) Heterogeneity in readouts of canonical wnt pathway activity within intestinal crypts. Dev. Dyn. Off. Publ. Am. Assoc. Anat. 245, 822–833

34 Li, N. et al. (2016) Heterogeneity in readouts of canonical wnt pathway activity within intestinal crypts. Dev. Dyn. Off. Publ. Am. Assoc. Anat. 245, 822–833

35 Lustig, B. et al. (2002) Negative feedback loop of Wnt signaling through upregulation of conductin/axin2 in colorectal and liver tumors. Mol. Cell. Biol. 22, 1184–1193

36 Durand, A. et al. (2012) Functional intestinal stem cells after Paneth cell ablation induced by the loss of transcription factor Math1 (Atoh1). Proc. Natl. Acad. Sci. U. S. A. 109, 8965–8970

37 van Es, J.H. et al. (2012) A critical role for the Wnt effector Tcf4 in adult intestinal homeostatic self-renewal. Mol. Cell. Biol. 32, 1918–1927

38 Tetteh, P.W. et al. (2016) Replacement of Lost Lgr5-Positive Stem Cells through Plasticity of Their Enterocyte-Lineage Daughters. Cell Stem Cell 18, 203–213

39 Narisawa, S. et al. (2003) Accelerated fat absorption in intestinal alkaline phosphatase knockout mice. Mol. Cell. Biol. 23, 7525–7530

40 Buczacki, S.J.A. et al. (2013) Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 495, 65–69