The gastrointestinal epithelium is composed of a cell lining that renews every 4–7 days, forming a barrier between the systemic circulation of the organism and the lumen of the intestine.
1- van der Flier L.G.
- Clevers H.
Stem cells, self-renewal, and differentiation in the intestinal epithelium.
In homeostasis, the rapid renewal of the epithelium originates from active intestinal stem cells (aISCs), which exist at the base of the crypt intercalated between Paneth cells.
2- 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.
- Clevers H.
Identification of stem cells in small intestine and colon by marker gene Lgr5.
These cells are marked by
Lgr5, which encodes a G protein–coupled receptor for R-spondins, and
Olfm4, a Notch target gene, with high fidelity within the murine and human small intestine.
2- 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.
- Clevers H.
Identification of stem cells in small intestine and colon by marker gene Lgr5.
, 3- Schuijers J.
- van der Flier L.G.
- van Es J.
- Clevers H.
Robust Cre-mediated recombination in small intestinal stem cells utilizing the Olfm4 locus.
, 4- VanDussen K.L.
- Carulli A.J.
- Keeley T.M.
- Patel S.R.
- Puthoff B.J.
- Magness S.T.
- Tran I.T.
- Maillard I.
- Siebel C.
- Å Kolterud
- Grosse A.S.
- Gumucio D.L.
- Ernst S.A.
- Tsai Y.-H.
- Dempsey P.J.
- Samuelson L.C.
Notch signaling modulates proliferation and differentiation of intestinal crypt base columnar stem cells.
These crypt-base stem cells are highly susceptible to irradiation and chemotherapy-induced injury.
5Extreme sensitivity of some intestinal crypt cells to X and γ irradiation.
, 6- Dekaney C.M.
- Gulati A.S.
- Garrison A.P.
- Helmrath M.A.
- Henning S.J.
Regeneration of intestinal stem/progenitor cells following doxorubicin treatment of mice.
, 7- Pritchard D.M.
- Watson A.J.M.
- Potten C.S.
- Jackman A.L.
- Hickman J.A.
Inhibition by uridine but not thymidine of p53-dependent intestinal apoptosis initiated by 5-fluorouracil: evidence for the involvement of RNA perturbation.
After acute injury to aISCs by irradiation, a reserve population of quiescent stem cells, labeled by
Bmi1 or
Hopx near the +4 crypt position, regenerate the epithelium.
8- Sangiorgi E.
- Capecchi M.R.
Bmi1 is expressed in vivo in intestinal stem cells.
, 9- Takeda N.
- Jain R.
- LeBoeuf M.R.
- Wang Q.
- Lu M.M.
- Epstein J.A.
Interconversion between intestinal stem cell populations in distinct niches.
, 10- Yan K.S.
- Chia L.A.
- Li X.
- Ootani A.
- Su J.
- Lee J.Y.
- Su N.
- Luo Y.
- Heilshorn S.C.
- Amieva M.R.
- Sangiorgi E.
- Capecchi M.R.
- Kuo C.J.
The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations.
Recent studies demonstrate that
Bmi1 marks cells of the enteroendocrine lineage.
11- Yan K.S.
- Gevaert O.
- Zheng G.X.Y.
- Anchang B.
- Probert C.S.
- Larkin K.A.
- Davies P.S.
- Cheng Z.
- Kaddis J.S.
- Han A.
- Roelf K.
- Calderon R.I.
- Cynn E.
- Hu X.
- Mandleywala K.
- Wilhelmy J.
- Grimes S.M.
- Corney D.C.
- Boutet S.C.
- Terry J.M.
- Belgrader P.
- Ziraldo S.B.
- Mikkelsen T.S.
- Wang F.
- Furstenberg RJ von
- Smith N.R.
- Chandrakesan P.
- May R.
- Chrissy M.A.S.
- Jain R.
- Cartwright C.A.
- Niland J.C.
- Hong Y.-K.
- Carrington J.
- Breault D.T.
- Epstein J.
- Houchen C.W.
- Lynch J.P.
- Martin M.G.
- Plevritis S.K.
- Curtis C.
- Ji H.P.
- Li L.
- Henning S.J.
- Wong M.H.
- Kuo C.J.
Intestinal enteroendocrine lineage cells possess homeostatic and injury-inducible stem cell activity.
Others have demonstrated the inherent plasticity of the intestinal epithelium, with varying degrees of regeneration originating from
Dll1+ or
Atoh1+ secretory progenitors,
12- van Es J.H.
- Sato T.
- van de Wetering M.
- Lyubimova A.
- Yee Nee A.N.
- Gregorieff A.
- Sasaki N.
- Zeinstra L.
- van den Born M.
- Korving J.
- Martens A.C.M.
- Barker N.
- van Oudenaarden A.
- Clevers H.
Dll1+ secretory progenitor cells revert to stem cells upon crypt damage.
, 13- Tomic G.
- Morrissey E.
- Kozar S.
- Ben-Moshe S.
- Hoyle A.
- Azzarelli R.
- Kemp R.
- Chilamakuri C.S.R.
- Itzkovitz S.
- Philpott A.
- Winton D.J.
Phospho-regulation of ATOH1 is required for plasticity of secretory progenitors and tissue regeneration.
, 14- Castillo-Azofeifa D.
- Fazio E.N.
- Nattiv R.
- Good H.J.
- Wald T.
- Pest M.A.
- de Sauvage F.J.
- Klein O.D.
- Asfaha S.
Atoh1+ secretory progenitors possess renewal capacity independent of Lgr5+ cells during colonic regeneration.
, 15- Bohin N.
- Keeley T.M.
- Carulli A.J.
- Walker E.M.
- Carlson E.A.
- Gao J.
- Aifantis I.
- Siebel C.W.
- Rajala M.W.
- Myers M.G.
- Jones J.C.
- Brindley C.D.
- Dempsey P.J.
- Samuelson L.C.
Rapid crypt cell remodeling regenerates the intestinal stem cell niche after notch inhibition.
differentiated
Alpi1+ enterocyte precursors,
16- Tetteh P.W.
- Basak O.
- Farin H.F.
- Wiebrands K.
- Kretzschmar K.
- Begthel H.
- van den Born M.
- Korving J.
- de Sauvage F.
- van Es J.H.
- van Oudenaarden A.
- Clevers H.
Replacement of lost Lgr5-positive stem cells through plasticity of their enterocyte-lineage daughters.
and differentiated
Defa4+,
Lyz1+, or
Bhlha15+ Paneth cells.
17- Jones J.C.
- Brindley C.D.
- Elder N.H.
- Myers M.G.
- Rajala M.W.
- Dekaney C.M.
- McNamee E.N.
- Frey M.R.
- Shroyer N.F.
- Dempsey P.J.
Cellular plasticity of Defa4Cre-expressing Paneth cells in response to notch activation and intestinal injury.
, 18- Yu S.
- Tong K.
- Zhao Y.
- Balasubramanian I.
- Yap G.S.
- Ferraris R.P.
- Bonder E.M.
- Verzi M.P.
- Gao N.
Paneth cell multipotency induced by notch activation following injury.
, 19- Hayakawa Y.
- Tsuboi M.
- Asfaha S.
- Kinoshita H.
- Niikura R.
- Konishi M.
- Hata M.
- Oya Y.
- Kim W.
- Middelhoff M.
- Hikiba Y.
- Higashijima N.
- Ihara S.
- Ushiku T.
- Fukayama M.
- Tailor Y.
- Hirata Y.
- Guha C.
- Yan K.S.
- Koike K.
- Wang T.C.
BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice.
Doxorubicin (DXR), a chemotherapy drug, and irradiation (IR) are considered to be similar injury models,
20Response of intestinal cells of differing topographical and hierarchical status to ten cytotoxic drugs and five sources of radiation.
, 21Further studies on the response of intestinal crypt cells of different hierarchical status to eighteen different cytotoxic agents.
, 22- Zhan Y.
- Xu C.
- Liu Z.
- Yang Y.
- Tan S.
- Yang Y.
- Jiang J.
- Liu H.
- Chen J.
- Wu B.
β -Arrestin1 inhibits chemotherapy-induced intestinal stem cell apoptosis and mucositis.
although the regenerative capacity of non–stem cells with DXR has not been fully explored.
6- Dekaney C.M.
- Gulati A.S.
- Garrison A.P.
- Helmrath M.A.
- Henning S.J.
Regeneration of intestinal stem/progenitor cells following doxorubicin treatment of mice.
,19- Hayakawa Y.
- Tsuboi M.
- Asfaha S.
- Kinoshita H.
- Niikura R.
- Konishi M.
- Hata M.
- Oya Y.
- Kim W.
- Middelhoff M.
- Hikiba Y.
- Higashijima N.
- Ihara S.
- Ushiku T.
- Fukayama M.
- Tailor Y.
- Hirata Y.
- Guha C.
- Yan K.S.
- Koike K.
- Wang T.C.
BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice.
Hayakawa et al
19- Hayakawa Y.
- Tsuboi M.
- Asfaha S.
- Kinoshita H.
- Niikura R.
- Konishi M.
- Hata M.
- Oya Y.
- Kim W.
- Middelhoff M.
- Hikiba Y.
- Higashijima N.
- Ihara S.
- Ushiku T.
- Fukayama M.
- Tailor Y.
- Hirata Y.
- Guha C.
- Yan K.S.
- Koike K.
- Wang T.C.
BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice.
demonstrated a reduction of aISCs within 24 hours after 15 mg/kg DXR. Despite this decrease in aISCs, lineage tracing from the
Lgr5 locus performed at the time of injury did not differ from noninjured intestines, indicating that the majority of crypts retained sufficient aISCs to repopulate the epithelium at this dose. Our previously published study demonstrated that there was an expansion of putative +4 or quiescent stem cells during the regenerative phase after DXR, although no transgenic mouse models were used to identify these cells, thus the findings required further investigation.
6- Dekaney C.M.
- Gulati A.S.
- Garrison A.P.
- Helmrath M.A.
- Henning S.J.
Regeneration of intestinal stem/progenitor cells following doxorubicin treatment of mice.
Secretory precursors and differentiated Paneth cells have a modest but variable (0–50 lineage trace events per 5 cm)
17- Jones J.C.
- Brindley C.D.
- Elder N.H.
- Myers M.G.
- Rajala M.W.
- Dekaney C.M.
- McNamee E.N.
- Frey M.R.
- Shroyer N.F.
- Dempsey P.J.
Cellular plasticity of Defa4Cre-expressing Paneth cells in response to notch activation and intestinal injury.
contribution to epithelial regeneration that has been identified in multiple studies.
15- Bohin N.
- Keeley T.M.
- Carulli A.J.
- Walker E.M.
- Carlson E.A.
- Gao J.
- Aifantis I.
- Siebel C.W.
- Rajala M.W.
- Myers M.G.
- Jones J.C.
- Brindley C.D.
- Dempsey P.J.
- Samuelson L.C.
Rapid crypt cell remodeling regenerates the intestinal stem cell niche after notch inhibition.
,17- Jones J.C.
- Brindley C.D.
- Elder N.H.
- Myers M.G.
- Rajala M.W.
- Dekaney C.M.
- McNamee E.N.
- Frey M.R.
- Shroyer N.F.
- Dempsey P.J.
Cellular plasticity of Defa4Cre-expressing Paneth cells in response to notch activation and intestinal injury.
,19- Hayakawa Y.
- Tsuboi M.
- Asfaha S.
- Kinoshita H.
- Niikura R.
- Konishi M.
- Hata M.
- Oya Y.
- Kim W.
- Middelhoff M.
- Hikiba Y.
- Higashijima N.
- Ihara S.
- Ushiku T.
- Fukayama M.
- Tailor Y.
- Hirata Y.
- Guha C.
- Yan K.S.
- Koike K.
- Wang T.C.
BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice.
,23- Buczacki S.J.A.
- Zecchini H.I.
- Nicholson A.M.
- Russell R.
- Vermeulen L.
- Kemp R.
- Winton D.J.
Intestinal label-retaining cells are secretory precursors expressing Lgr5.
This reversion phenomenon appears dependent on Notch activation after DXR and IR.
17- Jones J.C.
- Brindley C.D.
- Elder N.H.
- Myers M.G.
- Rajala M.W.
- Dekaney C.M.
- McNamee E.N.
- Frey M.R.
- Shroyer N.F.
- Dempsey P.J.
Cellular plasticity of Defa4Cre-expressing Paneth cells in response to notch activation and intestinal injury.
, 18- Yu S.
- Tong K.
- Zhao Y.
- Balasubramanian I.
- Yap G.S.
- Ferraris R.P.
- Bonder E.M.
- Verzi M.P.
- Gao N.
Paneth cell multipotency induced by notch activation following injury.
, 19- Hayakawa Y.
- Tsuboi M.
- Asfaha S.
- Kinoshita H.
- Niikura R.
- Konishi M.
- Hata M.
- Oya Y.
- Kim W.
- Middelhoff M.
- Hikiba Y.
- Higashijima N.
- Ihara S.
- Ushiku T.
- Fukayama M.
- Tailor Y.
- Hirata Y.
- Guha C.
- Yan K.S.
- Koike K.
- Wang T.C.
BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice.
Here, we show that DXR results in depletion of aISCs. After DXR-induced loss of aISCs, early progeny cells contribute to epithelial regeneration. However, after IR-induced loss of aISCs, there was not a similar recruitment of early progeny cells for regeneration, suggesting damage-dependent differences. After DXR, early progeny cells rarely underwent apoptosis, in contrast to the highly apoptotic aISC population. In homeostasis, early progeny cells are actively cycling, similar to aISCs. Therefore, quiescence does not appear to be a mechanism by which these cells are chemoresistant. Early progeny cells in homeostasis have decreased expression of DNA damage response transcripts in comparison with aISCs. A dampened DNA damage response may underlie the enhanced survival of early progeny cells in the face of chemotherapy. These data are supportive of early progeny cells as a highly flexible population with substantial capacity for regeneration after loss of aISCs.
Discussion
In this study, we demonstrate that a major source of epithelial regeneration after DXR-induced aISC depletion originates from early progeny cells that have recently exited the crypt base and no longer express stem cell–specific transcripts. These early progeny cells survived DXR-mediated apoptosis and began to express stem cell transcripts, as early as 6 and 24 hours after damage. DXR rapidly depleted aISCs via induction of p53-mediated cleavage of caspase-3, resulting in high levels of apoptosis in the aISCs. While we had previously suggested no reduction in the aISC population following DXR treatment,
6- Dekaney C.M.
- Gulati A.S.
- Garrison A.P.
- Helmrath M.A.
- Henning S.J.
Regeneration of intestinal stem/progenitor cells following doxorubicin treatment of mice.
in retrospect, this was not quantitatively assessed on a per crypt basis, and likely did not capture the extent of aISC depletion that is progressive over the first few days after injury.
Owing to their role as long lived highly proliferative cells, aISCs have a high commitment to genomic integrity.
58Balancing self-renewal against genome preservation in stem cells: how to have the cake and eat it too?.
When encountering high levels of DNA damage, these cells choose to undergo apoptosis, rather than perpetuate DNA mutations. DXR is a potent anthracycline chemotherapeutic with problematic off-target effects including cardiotoxicity, bone marrow suppression, and oral and intestinal mucositis. DXR undergoes rapid translocation to the nucleus, where it intercalates with nuclear and mitochondrial DNA, poisons topoisomerase II alpha, and produces free radicals.
56- Burgess D.J.
- Doles J.
- Zender L.
- Xue W.
- Ma B.
- McCombie W.R.
- Hannon G.J.
- Lowe S.W.
- Hemann M.T.
Topoisomerase levels determine chemotherapy response in vitro and in vivo.
,59- Yang F.
- Teves S.S.
- Kemp C.J.
- Henikoff S.
Doxorubicin, DNA torsion, and chromatin dynamics.
Consequently, we found that the majority of CASP3
+ cells in the crypt at 6 hours after DXR injection are aISCs. This is similar to previous findings after IR injury, in which 40% of apoptotic cells were
Lgr5eGFP+.
60- Zhu Y.
- Huang Y.-F.
- Kek C.
- Bulavin D.V.
Apoptosis differently affects lineage tracing of Lgr5 and Bmi1 intestinal stem cell populations.
We also observed expulsion of aISCs from enteroid buds in vitro after DXR. This is consistent with Andrade and Rosenblatt,
61Apoptotic regulation of epithelial cellular extrusion.
in which apoptosis but not necrosis was the inciting event for cellular extrusion from an intestinal epithelial monolayer. We have demonstrated that DXR results in a rapid, persistent loss of
Lgr5-expressing aISCs out to at least 5 days after insult. This was supported by the lack of lineage tracing when labeled was performed simultaneously with injury.
Although the regenerative response to IR has been extensively studied,
9- Takeda N.
- Jain R.
- LeBoeuf M.R.
- Wang Q.
- Lu M.M.
- Epstein J.A.
Interconversion between intestinal stem cell populations in distinct niches.
, 10- Yan K.S.
- Chia L.A.
- Li X.
- Ootani A.
- Su J.
- Lee J.Y.
- Su N.
- Luo Y.
- Heilshorn S.C.
- Amieva M.R.
- Sangiorgi E.
- Capecchi M.R.
- Kuo C.J.
The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations.
, 11- Yan K.S.
- Gevaert O.
- Zheng G.X.Y.
- Anchang B.
- Probert C.S.
- Larkin K.A.
- Davies P.S.
- Cheng Z.
- Kaddis J.S.
- Han A.
- Roelf K.
- Calderon R.I.
- Cynn E.
- Hu X.
- Mandleywala K.
- Wilhelmy J.
- Grimes S.M.
- Corney D.C.
- Boutet S.C.
- Terry J.M.
- Belgrader P.
- Ziraldo S.B.
- Mikkelsen T.S.
- Wang F.
- Furstenberg RJ von
- Smith N.R.
- Chandrakesan P.
- May R.
- Chrissy M.A.S.
- Jain R.
- Cartwright C.A.
- Niland J.C.
- Hong Y.-K.
- Carrington J.
- Breault D.T.
- Epstein J.
- Houchen C.W.
- Lynch J.P.
- Martin M.G.
- Plevritis S.K.
- Curtis C.
- Ji H.P.
- Li L.
- Henning S.J.
- Wong M.H.
- Kuo C.J.
Intestinal enteroendocrine lineage cells possess homeostatic and injury-inducible stem cell activity.
, 12- van Es J.H.
- Sato T.
- van de Wetering M.
- Lyubimova A.
- Yee Nee A.N.
- Gregorieff A.
- Sasaki N.
- Zeinstra L.
- van den Born M.
- Korving J.
- Martens A.C.M.
- Barker N.
- van Oudenaarden A.
- Clevers H.
Dll1+ secretory progenitor cells revert to stem cells upon crypt damage.
,15- Bohin N.
- Keeley T.M.
- Carulli A.J.
- Walker E.M.
- Carlson E.A.
- Gao J.
- Aifantis I.
- Siebel C.W.
- Rajala M.W.
- Myers M.G.
- Jones J.C.
- Brindley C.D.
- Dempsey P.J.
- Samuelson L.C.
Rapid crypt cell remodeling regenerates the intestinal stem cell niche after notch inhibition.
,18- Yu S.
- Tong K.
- Zhao Y.
- Balasubramanian I.
- Yap G.S.
- Ferraris R.P.
- Bonder E.M.
- Verzi M.P.
- Gao N.
Paneth cell multipotency induced by notch activation following injury.
,19- Hayakawa Y.
- Tsuboi M.
- Asfaha S.
- Kinoshita H.
- Niikura R.
- Konishi M.
- Hata M.
- Oya Y.
- Kim W.
- Middelhoff M.
- Hikiba Y.
- Higashijima N.
- Ihara S.
- Ushiku T.
- Fukayama M.
- Tailor Y.
- Hirata Y.
- Guha C.
- Yan K.S.
- Koike K.
- Wang T.C.
BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice.
,23- Buczacki S.J.A.
- Zecchini H.I.
- Nicholson A.M.
- Russell R.
- Vermeulen L.
- Kemp R.
- Winton D.J.
Intestinal label-retaining cells are secretory precursors expressing Lgr5.
,36- Powell A.E.
- Wang Y.
- Li Y.
- Poulin E.J.
- means A.L.
- Washington M.K.
- Higginbotham J.N.
- Juchheim A.
- Prasad N.
- Levy S.E.
- Guo Y.
- Shyr Y.
- Aronow B.J.
- Haigis K.M.
- Franklin J.L.
- Coffey R.J.
The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor.
,42- Barriga F.M.
- Montagni E.
- Mana M.
- Mendez-Lago M.
- Hernando-Momblona X.
- Sevillano M.
- Guillaumet-Adkins A.
- Rodriguez-Esteban G.
- Buczacki S.J.A.
- Gut M.
- Heyn H.
- Winton D.J.
- Yilmaz O.H.
- Attolini C.S.-O.
- Gut I.
- Batlle E.
Mex3a marks a slowly dividing subpopulation of Lgr5+ intestinal stem cells.
,48- Ayyaz A.
- Kumar S.
- Sangiorgi B.
- Ghoshal B.
- Gosio J.
- Ouladan S.
- Fink M.
- Barutcu S.
- Trcka D.
- Shen J.
- Chan K.
- Wrana J.L.
- Gregorieff A.
Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell.
less is understood about regeneration after DXR.
17- Jones J.C.
- Brindley C.D.
- Elder N.H.
- Myers M.G.
- Rajala M.W.
- Dekaney C.M.
- McNamee E.N.
- Frey M.R.
- Shroyer N.F.
- Dempsey P.J.
Cellular plasticity of Defa4Cre-expressing Paneth cells in response to notch activation and intestinal injury.
,19- Hayakawa Y.
- Tsuboi M.
- Asfaha S.
- Kinoshita H.
- Niikura R.
- Konishi M.
- Hata M.
- Oya Y.
- Kim W.
- Middelhoff M.
- Hikiba Y.
- Higashijima N.
- Ihara S.
- Ushiku T.
- Fukayama M.
- Tailor Y.
- Hirata Y.
- Guha C.
- Yan K.S.
- Koike K.
- Wang T.C.
BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice.
,23- Buczacki S.J.A.
- Zecchini H.I.
- Nicholson A.M.
- Russell R.
- Vermeulen L.
- Kemp R.
- Winton D.J.
Intestinal label-retaining cells are secretory precursors expressing Lgr5.
We were unable to identify contribution from
Hopx+ and
Bmi1+-expressing cells after DXR. Populations expressing these transcripts have exhibited an expanded capacity for regeneration after damage.
8- Sangiorgi E.
- Capecchi M.R.
Bmi1 is expressed in vivo in intestinal stem cells.
, 9- Takeda N.
- Jain R.
- LeBoeuf M.R.
- Wang Q.
- Lu M.M.
- Epstein J.A.
Interconversion between intestinal stem cell populations in distinct niches.
, 10- Yan K.S.
- Chia L.A.
- Li X.
- Ootani A.
- Su J.
- Lee J.Y.
- Su N.
- Luo Y.
- Heilshorn S.C.
- Amieva M.R.
- Sangiorgi E.
- Capecchi M.R.
- Kuo C.J.
The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations.
,37- Tian H.
- Biehs B.
- Warming S.
- Leong K.G.
- Rangell L.
- Klein O.D.
- de Sauvage F.J.
A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable.
These populations likely overlap with other crypt epithelial populations, as many of the proposed +4 markers are also expressed in active intestinal stem cells and
Bmi1+ also labels enteroendocrine lineage cells.
11- Yan K.S.
- Gevaert O.
- Zheng G.X.Y.
- Anchang B.
- Probert C.S.
- Larkin K.A.
- Davies P.S.
- Cheng Z.
- Kaddis J.S.
- Han A.
- Roelf K.
- Calderon R.I.
- Cynn E.
- Hu X.
- Mandleywala K.
- Wilhelmy J.
- Grimes S.M.
- Corney D.C.
- Boutet S.C.
- Terry J.M.
- Belgrader P.
- Ziraldo S.B.
- Mikkelsen T.S.
- Wang F.
- Furstenberg RJ von
- Smith N.R.
- Chandrakesan P.
- May R.
- Chrissy M.A.S.
- Jain R.
- Cartwright C.A.
- Niland J.C.
- Hong Y.-K.
- Carrington J.
- Breault D.T.
- Epstein J.
- Houchen C.W.
- Lynch J.P.
- Martin M.G.
- Plevritis S.K.
- Curtis C.
- Ji H.P.
- Li L.
- Henning S.J.
- Wong M.H.
- Kuo C.J.
Intestinal enteroendocrine lineage cells possess homeostatic and injury-inducible stem cell activity.
,44- Muñoz J.
- Stange D.E.
- Schepers A.G.
- van de Wetering M.
- Koo B.-K.
- Itzkovitz S.
- Volckmann R.
- Kung K.S.
- Koster J.
- Radulescu S.
- Myant K.
- Versteeg R.
- Sansom O.J.
- van Es J.H.
- Barker N.
- van Oudenaarden A.
- Mohammed S.
- Heck A.J.R.
- Clevers H.
The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent ‘+4’ cell markers.
,45Heterogeneity of the level of activity of lgr5+ intestinal stem cells.
Additionally,
Bmi1CreERT expression decreases from the duodenum to the ileum.
37- Tian H.
- Biehs B.
- Warming S.
- Leong K.G.
- Rangell L.
- Klein O.D.
- de Sauvage F.J.
A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable.
However, there was sufficient jejunal labeling present to monitor for any expansion in regenerative contribution. In future experiments, it would be interesting to explore the regenerative hierarchy in other regions of the intestine.
As the early progeny cells in this study appear at position +4–7, it is likely we are labeling TA cells that are not yet differentiated enough to robustly express lineage specific programs, such as enterocyte-specific
Alpi1.
16- Tetteh P.W.
- Basak O.
- Farin H.F.
- Wiebrands K.
- Kretzschmar K.
- Begthel H.
- van den Born M.
- Korving J.
- de Sauvage F.
- van Es J.H.
- van Oudenaarden A.
- Clevers H.
Replacement of lost Lgr5-positive stem cells through plasticity of their enterocyte-lineage daughters.
Given that the bulk of epithelial cells are enterocytes, and that secretory progenitors, labeled by
Dll1+, did not contribute to epithelial regeneration in our model, we speculate that a majority of early progeny cells are destined for enterocyte identities under homeostatic conditions. Secretory progenitors require Notch activation to be able to reacquire stem-like properties after DXR damage in other studies.
17- Jones J.C.
- Brindley C.D.
- Elder N.H.
- Myers M.G.
- Rajala M.W.
- Dekaney C.M.
- McNamee E.N.
- Frey M.R.
- Shroyer N.F.
- Dempsey P.J.
Cellular plasticity of Defa4Cre-expressing Paneth cells in response to notch activation and intestinal injury.
, 18- Yu S.
- Tong K.
- Zhao Y.
- Balasubramanian I.
- Yap G.S.
- Ferraris R.P.
- Bonder E.M.
- Verzi M.P.
- Gao N.
Paneth cell multipotency induced by notch activation following injury.
, 19- Hayakawa Y.
- Tsuboi M.
- Asfaha S.
- Kinoshita H.
- Niikura R.
- Konishi M.
- Hata M.
- Oya Y.
- Kim W.
- Middelhoff M.
- Hikiba Y.
- Higashijima N.
- Ihara S.
- Ushiku T.
- Fukayama M.
- Tailor Y.
- Hirata Y.
- Guha C.
- Yan K.S.
- Koike K.
- Wang T.C.
BHLHA15-positive secretory precursor cells can give rise to tumors in intestine and colon in mice.
It is possible there is a dose-dependent response to DXR that alters Notch programming, as we did not observe recruitment of secretory cells or their progenitors in response to the dose of DXR used in this study.
The significant contribution to regeneration by early progeny cells after DXR, but not IR, was unexpected. A recent publication demonstrated that a subpopulation of aISCs in the
Lgr5eGFP-CreERT2 mouse were retained after 10 Gy IR.
27- Sato T.
- Sase M.
- Ishikawa S.
- Kajita M.
- Asano J.
- Sato T.
- Mori Y.
- Ohteki T.
Characterization of radioresistant epithelial stem cell heterogeneity in the damaged mouse intestine.
While these cells downregulate GFP expression, they are able to produce significant percentages of lineage tracing postinjury. Other studies have used 12 Gy to induce crypt and aISC loss,
10- Yan K.S.
- Chia L.A.
- Li X.
- Ootani A.
- Su J.
- Lee J.Y.
- Su N.
- Luo Y.
- Heilshorn S.C.
- Amieva M.R.
- Sangiorgi E.
- Capecchi M.R.
- Kuo C.J.
The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations.
,42- Barriga F.M.
- Montagni E.
- Mana M.
- Mendez-Lago M.
- Hernando-Momblona X.
- Sevillano M.
- Guillaumet-Adkins A.
- Rodriguez-Esteban G.
- Buczacki S.J.A.
- Gut M.
- Heyn H.
- Winton D.J.
- Yilmaz O.H.
- Attolini C.S.-O.
- Gut I.
- Batlle E.
Mex3a marks a slowly dividing subpopulation of Lgr5+ intestinal stem cells.
,48- Ayyaz A.
- Kumar S.
- Sangiorgi B.
- Ghoshal B.
- Gosio J.
- Ouladan S.
- Fink M.
- Barutcu S.
- Trcka D.
- Shen J.
- Chan K.
- Wrana J.L.
- Gregorieff A.
Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell.
,54- Hua G.
- Thin T.H.
- Feldman R.
- Haimovitz-Friedman A.
- Clevers H.
- Fuks Z.
- Kolesnick R.
Crypt base columnar stem cells in small intestines of mice are radioresistant.
,62- Gregorieff A.
- Liu Y.
- Inanlou M.R.
- Khomchuk Y.
- Wrana J.L.
Yap-dependent reprogramming of Lgr5 + stem cells drives intestinal regeneration and cancer.
,63Protection of the small intestinal clonogenic stem cells from radiation-induced damage by pretreatment with interleukin 11 also increases murine survival time.
and in this study we demonstrate reduced lineage tracing from aISCs at 12 Gy, suggestive of aISC depletion. Thus, the difference in regenerative contribution after IR between Sato et al
27- Sato T.
- Sase M.
- Ishikawa S.
- Kajita M.
- Asano J.
- Sato T.
- Mori Y.
- Ohteki T.
Characterization of radioresistant epithelial stem cell heterogeneity in the damaged mouse intestine.
and the data shown here may be due to severity of the injury. Ayyaz et al
48- Ayyaz A.
- Kumar S.
- Sangiorgi B.
- Ghoshal B.
- Gosio J.
- Ouladan S.
- Fink M.
- Barutcu S.
- Trcka D.
- Shen J.
- Chan K.
- Wrana J.L.
- Gregorieff A.
Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell.
utilized single cell transcriptomics to identify a revival stem cell population which has regenerative potential in the face of IR. These cells arise infrequently from
Lgr5+ cells, tend to be quiescent, and take up to a week after induction of labeling to appear.
48- Ayyaz A.
- Kumar S.
- Sangiorgi B.
- Ghoshal B.
- Gosio J.
- Ouladan S.
- Fink M.
- Barutcu S.
- Trcka D.
- Shen J.
- Chan K.
- Wrana J.L.
- Gregorieff A.
Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell.
Similarly, a recent study by Murata et al
49- Murata K.
- Jadhav U.
- Madha S.
- van Es J.
- Dean J.
- Cavazza A.
- Wucherpfennig K.
- Michor F.
- Clevers H.
- Shivdasani R.A.
Ascl2-dependent cell dedifferentiation drives regeneration of ablated intestinal stem cells.
demonstrated that aISC progeny generated within 4 days prior to IR contribute substantially to epithelial regeneration. The dedifferentiation potential of the progeny cells was dependent on the transcription factor
Ascl2.
49- Murata K.
- Jadhav U.
- Madha S.
- van Es J.
- Dean J.
- Cavazza A.
- Wucherpfennig K.
- Michor F.
- Clevers H.
- Shivdasani R.A.
Ascl2-dependent cell dedifferentiation drives regeneration of ablated intestinal stem cells.
The overlap of the previous studies with the early progeny cells identified here is unknown. Our study indicates that substantive repair after aISC depletion by DXR, but not IR, can be performed by cells generated within a day from
Lgr5+ aISCs. The 1-day duration of labeling in our study is more restrictive than that of Murata et al’s
49- Murata K.
- Jadhav U.
- Madha S.
- van Es J.
- Dean J.
- Cavazza A.
- Wucherpfennig K.
- Michor F.
- Clevers H.
- Shivdasani R.A.
Ascl2-dependent cell dedifferentiation drives regeneration of ablated intestinal stem cells.
experimental design. This suggests that early progeny cells are more sensitive to IR than progeny further away from the crypt base. However, we also found that early progeny cells expressed higher levels of
Ascl2 after injury, suggesting a possible common regenerative pathway between damage models.
49- Murata K.
- Jadhav U.
- Madha S.
- van Es J.
- Dean J.
- Cavazza A.
- Wucherpfennig K.
- Michor F.
- Clevers H.
- Shivdasani R.A.
Ascl2-dependent cell dedifferentiation drives regeneration of ablated intestinal stem cells.
,64Ascl2 reinforces intestinal stem cell identity.
Taken together, these studies and our current body of work suggest that the TA population exhibits a high degree of plasticity in response to crypt injury.
Why do these early progeny cells survive DXR damage? Quiescence does not appear to be a major chemoresistant mechanism for these cells, as the percentage of early progeny cells incorporating BrdU was similar to double
+ aISCs, and significantly more than GFP-only aISCs. It also seems surprising to us that cells that share a close spatial relationship, such as early progeny cells and aISCs, would have such disparate responses to DNA damage. Tao et al
43- Tao S.
- Tang D.
- Morita Y.
- Sperka T.
- Omrani O.
- Lechel A.
- Sakk V.
- Kraus J.
- Kestler H.A.
- Kühl M.
- Rudolph K.L.
Wnt activity and basal niche position sensitize intestinal stem and progenitor cells to DNA damage.
demonstrated that the Wnt signaling gradient is an important factor in sensitivity to DNA damage, in which cells that are further from the crypt base are less sensitive. Although that study was limited to
Lgr5eGFP+ cells, it seems plausible that differences in the strength of Wnt signaling could drive the balance between chemosensitivity and resistance in our model.
We have identified differences between early progeny cells and aISCs with regard to expression of DDR transcripts.
Atm, a mechanism by which cells sense and signal DNA breaks, is not expressed differently in our isolated populations. However, the early progeny cells do have reduced expression of transcripts encoding proteins that are involved in homologous recombination (HR). HR is critical for accurately repairing double-stranded DNA breaks. It seems counterintuitive that early progeny cells, which are able to survive DXR, would have reduced transcript levels of HR-associated genes. However, we speculate that the reduced expression of these DDR genes decreases predisposition to intrinsic apoptosis. In support of this hypothesis, Watanabe et al
55- Watanabe K.
- Ikuno Y.
- Kakeya Y.
- Ikeno S.
- Taniura H.
- Kurono M.
- Minemori K.
- Katsuyama Y.
- Naka-Kaneda H.
Age-related dysfunction of the DNA damage response in intestinal stem cells.
identified that reduced DDR is associated with reduced apoptosis, marked by CASP3, in aging ISC populations. Mutations in surviving and proliferating cells after DNA damaging insults could play a role in cancer initiation. We do not know at this time whether there is persistent DNA damage or mutations retained in the early progeny cells that make up the newly formed stem cell compartment after DXR-induced injury, and what impact this may have on potential development of preneoplastic or neoplastic lesions.
Finally, topoisomerase II alpha-DNA complexes, formed as TOP2A, introduces double-strand breaks after replication or to relieve stresses from coiled DNA, are stabilized by DXR.
56- Burgess D.J.
- Doles J.
- Zender L.
- Xue W.
- Ma B.
- McCombie W.R.
- Hannon G.J.
- Lowe S.W.
- Hemann M.T.
Topoisomerase levels determine chemotherapy response in vitro and in vivo.
This stabilization leads to irreversible DNA damage, driving cells toward apoptosis. Early progeny cells express modestly lower levels of
Top2a transcript as compared with aISCs, although whether this also relates to reduced TOP2A protein is unknown. If early progeny cells have decreased TOP2A, this could result in reduced poisoning of TOP2A-DNA complexes and therefore less irreversible DNA damage in early progeny cells than in aISCs. Thus, early progeny cells would have less sensitivity to DXR-induced damage. Further research is necessary to explore these hypotheses relating to chemoresistance in the intestinal epithelium.
Here, we have identified that early progeny cells of aISCs are a major contributing population to epithelial regeneration after chemotherapeutic insult to the small intestine. Rapid regeneration of the epithelium is critical to maintaining epithelial barrier function. Our study demonstrates that the highly flexible cellular identity of the crypt epithelium is critical to regeneration of the epithelium after chemotherapeutic injury. This finding could be harnessed to develop therapeutic strategies to minimize the severity of chemotherapy-associated mucositis and aISC damage.
Materials and Methods
Mice
Lgr5IRES-eGFP-CreERT2 (JAX stock # 008875; The Jackson Laboratory, Bar Harbor, ME)
2- 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.
- Clevers H.
Identification of stem cells in small intestine and colon by marker gene Lgr5.
and
Defa6iCre41- Adolph T.E.
- Tomczak M.F.
- Niederreiter L.
- Ko H.-J.
- Böck J.
- Martinez-Naves E.
- Glickman J.N.
- Tschurtschenthaler M.
- Hartwig J.
- Hosomi S.
- Flak M.B.
- Cusick J.L.
- Kohno K.
- Iwawaki T.
- Billmann-Born S.
- Raine T.
- Bharti R.
- Lucius R.
- Kweon M.-N.
- Marciniak S.J.
- Choi A.
- Hagen S.J.
- Schreiber S.
- Rosenstiel P.
- Kaser A.
- Blumberg R.S.
Paneth cells as a site of origin for intestinal inflammation.
mice were bred in-house at North Carolina State University from established lines (all on C57BL/6 background).
Defa6iCre mice were a gift from R. Blumberg at Brigham and Women’s Hospital (Boston, MA).
HopxCreERT2 (JAX stock #017606; The Jackson Laboratory)
9- Takeda N.
- Jain R.
- LeBoeuf M.R.
- Wang Q.
- Lu M.M.
- Epstein J.A.
Interconversion between intestinal stem cell populations in distinct niches.
and
Bmi1CreERT (JAX stock #010531; The Jackson Laboratory)
8- Sangiorgi E.
- Capecchi M.R.
Bmi1 is expressed in vivo in intestinal stem cells.
were obtained from JAX labs and maintained in-house.
Rosa26LSL-tdTomato mice (JAX stock # 007914; The Jackson Laboratory) were crossed with the above Cre lines to generate mice for lineage-tracing experiments (reporter mice). Unstained cells and single-color control populations for flow cytometry were obtained from the jejunal epithelia of wild-type C57BL/6 mice. All animals were cared for under the North Carolina State University’s Institutional Animal Care and Use Committee guidelines. Experiments at North Carolina State University were performed by B. Sheahan or A. Freeman. Experiments were performed on 8- to 20-week-old mice, with male and female mice randomly allocated to experimental groups. Control mice were injected with TAM at the indicated time points.
Dll1eGFP-CreERT212- van Es J.H.
- Sato T.
- van de Wetering M.
- Lyubimova A.
- Yee Nee A.N.
- Gregorieff A.
- Sasaki N.
- Zeinstra L.
- van den Born M.
- Korving J.
- Martens A.C.M.
- Barker N.
- van Oudenaarden A.
- Clevers H.
Dll1+ secretory progenitor cells revert to stem cells upon crypt damage.
and
Olfm4eGFP-CreERT23- Schuijers J.
- van der Flier L.G.
- van Es J.
- Clevers H.
Robust Cre-mediated recombination in small intestinal stem cells utilizing the Olfm4 locus.
mice were crossed with
Rosa26LSL-tdTomato mice (all on a C57BL/6 background) at University of Michigan to generate mice for lineage-tracing experiments. All animals were cared for under the University of Michigan’s Institutional Animal Care and Use Committee guidelines. Experiments at University of Michigan were performed by T. Keeley. Experiments were performed on 8- to 16-week-old mice, with male and female mice randomly allocated to experimental groups. Control mice were injected with TAM at the indicated time points.
Lgr5IRES-CreERT232- Huch M.
- Dorrell C.
- Boj S.F.
- van Es J.H.
- Li V.S.W.
- van de Wetering M.
- Sato T.
- Hamer K.
- Sasaki N.
- Finegold M.J.
- Haft A.
- Vries R.G.
- Grompe M.
- Clevers H.
In vitro expansion of single Lgr5 + liver stem cells induced by Wnt-driven regeneration.
mice were crossed with
Rosa26LSL-tdTomato mice (all on a C57BL/6 background) at Duke University to generate mice for lineage-tracing experiments. All animals were cared for under Duke University’s Institutional Animal Care and Use Committee guidelines. Tissues were collected by B. Sheahan. Experiments were performed on 27-week-old mice, with male and female mice randomly allocated to experimental groups. The same control mice were analyzed 5 days after TAM injection to quantify lineage tracing to compare to the 2 experimental groups.
Mice were kept in grouped housing and maintained under a 12-hour light/dark cycle and fed regular free-choice chow through all experimental procedures. When possible, mice of similar age and sex (littermate control animals) were used for experimental and control groups. The n for each experiment is indicated either in the figure legend or graphically represented by symbols.
Mice were euthanized by cervical dislocation after anesthesia with isoflurane. Small intestine was immediately collected after euthanasia and flushed with ice-cold 1× phosphate-buffered saline (PBS) (Ca2+ and Mg2+ free). The small intestine proximal to the ligament of Treitz (signifying the duodenojejunal juncture) was discarded, and the proximal one-half (approximately 10–12 cm) of the remaining intestine was identified as jejunum.
Treatments
DXR Injection
Mice were injected once with 20 mg/kg DXR HCl (Actavis, Parsippany-Troy Hills, NJ) intraperitoneally. Mice were monitored for weight loss daily. If mice lost >20% of initial body weight, they were euthanized in accordance with Institutional Animal Care and Use Committee protocols. Control mice did not receive a vehicle injection in lieu of DXR.
Total Body Irradiation
Mice were exposed to 12 Gy of total body irradiation (TBI). TBI was performed 50 cm from the radiation source with a dose rate of 3.18 Gy/min with 320 kVp x-rays, using 12.5 mA and a filter consisting of 2.5-mm Al and 0.1-mm Cu (X-RAD 320 Biological Irradiator; Precision X-Ray, North Branford, CT). TBI was performed by S. Hasapis. The dose rate was measured with an ion chamber by members of the Radiation Safety Division at Duke University. Control mice were not irradiated.
Labeling of Cells in S-Phase
Mice were injected with 150 mg/kg BrdU dissolved in sterile water intraperitoneally 2 hours prior to collection of small intestinal tissues.
Induction of Lineage Tracing
Mice were injected with 50 mg/kg TAM (Sigma-Aldrich, St Louis, MO) in corn oil (
Olfm4eGFP-CreERT2 and
Dll1eGFP-CreERT2 reporter mice) or sunflower seed oil (all mice at North Carolina State University and
Lgr5CreERT2 reporter mice) intraperitoneally at various time points prior to collection of small intestinal tissues. The TAM was reconstituted in 100% ethanol to 100 mg/mL, then diluted to 10 mg/mL with corn oil or sunflower seed oil prior to injection. We validated that all possible crypts were effectively labeled with this TAM dose using
Lgr5eGFP-CreERT2 reporter mice. In these mice, the number of GFP
+ crypts were not different from the number of tdTomato
+ crypts (
Figure 7B).
Quantitative RT-PCR
Total RNA was isolated from jejunal tissues with the RNeasy Mini kit (Qiagen, Hilden, Germany) per the manufacturer’s protocol. Quality of messenger RNA was verified with a Nanodrop 2000 spectrophotometer (Thermo Fisher Scientific, Waltham, MA). 500 ng cDNA was synthesized using the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA), with RNase A included in the reaction, following the manufacturer’s protocol. qRT-PCR analysis was performed with 25 ng complementary DNA/well using TaqMan Universal Master Mix II with UNG (Applied Biosystems), on a QuantStudio 6 PCR system (Thermo Fisher Scientific) for the following TaqMan probes: Actb (Mm02619580_g1), Lgr5 (Mm00438890_m1), Olfm4 (Mm01320260_m1), Hopx (Mm00558630_m1), and Bmi1 (Mm03053308_g1). All samples were run in triplicate. Signals were normalized to Actb for each sample, and relative fold changes were calculated via ΔΔCt analysis.
Crypt Culture for Time-Lapse Microscopy of GFP+ Cells
Chemical and mechanical dissociation was performed to obtain jejunal crypts as previously described with modifications.
65- 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.
- others
Single Lgr5 stem cells build crypt villus structures in vitro without a mesenchymal niche.
After filleting the length of the isolated jejunum, villi were removed by scraping the luminal side of the jejunum with a coverslip. The jejunal whole tissue was then incubated in 30 mM EDTA (pH 7.4) for 30 minutes on ice. Tissue was transferred to 1× PBS (Ca
2+ and Mg
2+ free), and mechanical dissociation (shaking) was performed to exfoliate crypts from the underlying lamina propria. Crypts were separated from intact villi by passage through a 70-μm cell strainer prior to counting. Approximately 100 isolated jejunal crypts were resuspended in 20 μL Matrigel (Corning, Corning, NY) and placed in 48-well tissue culture plates. After polymerization of the Matrigel, 250 μL of media was added per well. The media consisted of Advanced Dulbecco’s modified Eagle medium (DMEM)/F12 (Invitrogen, Carlsbad, CA) containing growth factors: 50 ng/mL recombinant mouse EGF (R&D Systems, Minneapolis, MN), 500 ng/mL R-spondin 1 (R&D Systems), 100 ng/mL recombinant mouse Noggin (PeproTech, Rocky Hill, NJ), 1× N2 supplement (Gibco, Gaithersburg, MD), 1× B27 (Gibco), 10 μM HEPES (Gibco), 1× Glutamax (Gibco), and 500 μg/mL penicillin-streptomycin (Gibco). Media were changed every other day. Enteroids were imaged daily for evidence of growth and budding. At 3 days postplating, 4 μL of DXR diluted in media was added to the media of treatment wells for a final concentration of 4 ng/μL. The same volume of fresh media was added to the media of control wells. GFP-expressing crypts were then monitored by time-lapse fluorescence microscopy (images obtained every 30 minutes for 12 hours after DXR addition) using a stage top incubator on the automated motorized stage of an inverted Olympus IX83 microscope (Olympus, Tokyo, Japan). Time-lapse images were processed with 2-dimensional deconvolution in CellSens (Olympus) prior to measurements. For each indicated time point, the distance (μm) of maximal GFP
+ intensity (representing
Lgr5eGFP+ cells) from the basolateral membrane of the enteroid bud was measured in the CellSens program (Olympus).
Transcriptional Analysis of Early Progeny Cells and aISCs
Prior to crypt-enriched epithelial cell isolation, 8- to 10-week-old Lgr5eGFP-CreERT2 reporter mice (n = 9) were injected with TAM. Control mice (n = 3) were analyzed 24 hours after TAM injection. DXR-treated mice were injected with DXR at 24 hours after TAM injection, then analyzed at either 6 hours (n = 3) or 24 hours (n = 3) after DXR. No mice were pooled for cell isolation.
Isolated jejunal crypts were centrifuged (1000 RPM × 5 minutes, 4°C) then resuspended in 1× Hank’s Balanced Salt Solution with 1 mg/mL dispase (Corning; 354235), and incubated at 37°C for 10 minutes with intermittent mechanical dissociation (shaking) to obtain a single cell suspension. After 10 minutes, 5 μg/mL of DNase I (Sigma-Aldrich), and 10% fetal bovine serum (FBS) (Gibco) were added to the suspension. The suspension was centrifuged (1000 RPM × 5 minutes, 4°C) and the pellet was resuspended in Advanced DMEM/F12 (Gibco) with 10% FBS and 5 μg/mL of DNase I. The cells were filtered through a 30-μm cell strainer prior to estimating the total number of cells per sample with a hemocytometer. The cells were centrifuged and resuspended at a concentration of 1 × 106 cells/100 μL in Advanced DMEM/F12 with 10% FBS and 5 μg/mL of DNase I. Cells were incubated with 2 μL 7-AAD (BioLegend, San Diego, CA; 420403) and 1 μL EPCAM-PE/Cy7 (BioLegend; 118215) per 1 × 106 cells for 20 minutes in the dark prior to sorting. Cells were kept on ice throughout antibody incubation and sorting.
FACS was performed with a Beckman Coulter MoFlow XDP cell sorter (Beckman Coulter, Brea, CA) in the Flow Cytometry and Cell Sorting Facility at North Carolina State University College of Veterinary Medicine. Events were plotted on forward scatter vs side scatter to exclude debris. Events were gated on side-scatter width vs side-scatter height to identify single-cell events. Single events were then gated for live epithelial cells, identified as 7-AAD negative and EPCAM-PE/Cy7 positive. Live epithelial cells were sorted on the basis of endogenous tdTomato and GFP fluorescence. Non–TAM-injected Lgr5eGFPCre-ERT2 reporter mice were examined to identify any background Cre activation. Background Cre activation in these reporter mice was minimal (0.03% tdTomato+ cells total in representative FACS plot). 500–30,000 cells per population of interest were sorted from each sample. Cells were sorted directly into Buffer RL (Norgen BioTek, Hamilton, Ontario, Canada; 17200) with 1% β-mercaptoethanol prior to freezing in liquid nitrogen and storage at –80°C. Total cells analyzed by the cell sorter ranged between 3.57–13.2 × 106/sample.
RNA was isolated from the sorted cell populations using Norgen Biotek RNA isolation kit (Norgen; 17200). 0.5 ng of RNA was transformed into complementary DNA using Fluidigm Reverse Transcription Master Mix (Fluidigm, South San Francisco, CA). One mouse (6 hours DXR group) was removed from analysis due to inadvertent TAM exposure from cagemate’s feces. At the Center for Gastroenterology Biology and Disease University of North Carolina Advanced Analytics Core facility, the complementary DNA samples underwent preamplification (Fluidigm) prior to loading on a 48.48 IFC plate for Ct analysis using the Fluidigm Biomark HD (Fluidigm) with TaqMan probes. Signals were normalized to
Actb and
Gapdh for each sample, and relative log2 fold changes were calculated, normalizing to the indicated Control transcript expressions for each panel in
Figures 8 and
10.
Immunofluorescence
For immunofluorescence, jejunal tissues were fixed in 4% paraformaldehyde for 12–18 hours, dehydrated in 30% sucrose for 24 hours, embedded in optimal cutting temperature compound, and stored at –80°C until sectioning. The 7-μm sections were adhered to Superfrost Plus slides (Thermo Fisher Scientific; 4951PLUS4) and OCT was removed by immersion in 1× PBS after drying. Sections were mounted with Hard Set mounting medium with DAPI (Vector Laboratories, Burlingame, CA; H-1500) and imaged with an inverted Olympus IX83 microscope. Control sections were incubated with IgG of the same primary species or blocking solution (5% bovine serum albumin or 10% FBS in PBS) for all immunofluorescent experiments.
For lysozyme and RFP immunofluorescence, sections were antigen retrieved in sodium citrate buffer (2.94 g sodium citrate, 300 μL Tween 80, pH 6) using a pressure cooker. Sections were blocked in 10% FBS in PBS for 1 hour at room temperature, incubated overnight at 4°C with primary antibodies, washed in 1× PBS, and incubated for 1 hour at room temperature with secondary antibodies. Sections were immersed in 1× PBS and mounted as described previously. Antibodies included: goat anti-lysozyme (1:500, sc27958), rabbit anti-RFP (1:250, R10367), anti-goat DyLight 488 (1:500, SA5-10086), and anti-rabbit AF555 (1:500, A21428).
For cleaved caspase-3, OLFM4, sucrase-isomaltase, CHGA, MUC2, or EPCAM immunofluorescence, sections were lightly fixed and permeabilized with methanol/acetone (50%/50%) for 20 minutes at –20°C. After washing with 1× PBS, sections were blocked with 5% bovine serum albumin in PBS for 1 hour at room temperature, incubated overnight at 4°C with primary antibodies, washed in 1× PBS, and incubated for 1 hour at room temperature with secondary antibodies. Sections were immersed in 1× PBS and mounted as described previously. Primary antibodies included: rabbit anti-CASP3 (1:250, 9579S), rabbit anti-OLFM4 (1:500, 39141S), goat anti-sucrase-isomaltase (1:250, sc27603), rabbit anti-CHGA (1:250, ab15160), rabbit anti-MUC2 (1:250, sc15334), and rabbit anti-EPCAM (1:500, ab71916). Secondary antibodies included: anti-rabbit DyLight 649 (1:500, 406406), anti-rabbit AF488 (1:500, A21206), and anti-goat AF 647 (1:500, A21244). We performed EPCAM immunofluorescence to validate the quantification of total epithelial crypts in tissues to ensure correct blinded counting of crypts using DAPI fluorescence. The total number of crypts counted with DAPI fluorescence and EPCAM fluorescence was not different.
For co-immunofluorescence of BrdU, GFP, and tdTomato, sections were antigen retrieved in sodium citrate buffer as previous, then permeabilized with 0.3% Triton X-100 for 10 minutes, washed in 1× PBS, and blocked with 10% FBS in PBS for 1 hour at room temperature. Sections were incubated with rabbit anti-GFP (1:2000, ab183734) in Signal Stain Antibody Diluent (Cell Signaling Technology, Danvers, MA; #8112) for 1 hour at room temperature, followed by Signal Stain Boost (Cell Signaling Technology; #8114) for 30 minutes at room temperature. Fluorescein reagent (PerkinElmer, Waltham, MA; NEL741001KT) was applied to the sections for 10 minutes prior to the stripping procedure. Slides were boiled in citrate stripping solution (10 mM sodium citrate, pH 6) and held at a sub-boiling temperature (90°C) for 10 minutes then cooled to room temperature. Following stripping, rabbit anti-RFP (1:500, R10367) and rat anti-BrdU (1:250, NB-500-169) were applied to the sections overnight at 4°C. After washing in 1× PBS, the following secondary antibodies were applied for 1 hour at room temperature: anti-rabbit AF555 (1:500, A-21428) and anti-rat APC (1:500, A10540).
Lineage-Tracing Assessment
At least 200 crypts were counted per mouse for Lgr5eGFP-CreERT2, Lgr5CreERT2, Defa6iCre, HopxCreERT2 and Bmi1CreERT reporter mice. At least 50 crypts were counted per mouse for Dll1eGFP-CreERT2 and Olfm4eGFP-CreERT2 reporter mice. Total crypts were enumerated using DAPI fluorescence. tdTomato+ lineage-tracing events were considered positive if >5 contiguous tdTomato+ cells were emanating from a crypt base. Events limited to villi or unconnected to a crypt were not considered positive.
OLFM4 Scoring
OLFM4 is secreted apically into the lumen of the small intestine in mice, making it difficult to identify which cells are specifically positive for this protein.
66- van der Flier LG.
- Haegebarth A.
- Stange D.E.
- van de Wetering M.
- Clevers H.
OLFM4 is a robust marker for stem cells in human intestine and marks a subset of colorectal cancer cells.
A blinded scoring system was adapted from Besson et al
67- Besson D.
- Pavageau A.-H.
- Valo I.
- Bourreau A.
- Bélanger A.
- Eymerit-Morin C.
- Moulière A.
- Chassevent A.
- Boisdron-Celle M.
- Morel A.
- Solassol J.
- Campone M.
- Gamelin E.
- Barré B.
- Coqueret O.
- Guette C.
A quantitative proteomic approach of the different stages of colorectal cancer establishes OLFM4 as a new nonmetastatic tumor marker.
to capture 4 categories of OLFM4 immunopositivity. These were scored by the intensity of the fluorescence to approximate the number of cells positive for OLFM4. Only complete jejunal hemicrypts were scored (>10 crypts/mouse). The scoring scheme is as follows: 0 = negative; 1 = 1–2 positive cells/faint fluorescence; 2 = 3–5 positive cells/moderate fluorescence; 3 = 6+ positive cells/intense fluorescence.
In Situ Hybridization
For in situ hybridization, tissues were fixed in 10% zinc formalin for 12–18 hours and moved to 70% ethanol before embedding in paraffin. Then, 5 μm sections were adhered to Superfrost Plus slides (Thermo Fisher Scientific; 4951PLUS4). Sections underwent standard deparaffinization with Histo Clear II (Thermo Fisher Scientific; 50-899-90150). In situ hybridization for Olfm4 probe was performed using RNAscope chromogenic assay 2.5, with Ppib as positive control probe and Dapb as negative control probe according to manufacturer’s instructions (Advanced Cell Diagnostics, Newark, CA). Only epithelial crypt cells were considered to be cells of interest.
Statistical Analysis
All statistics and preparation of graphs were performed in GraphPad 8 (GraphPad Software, San Diego, CA). FIJI was utilized for image analysis and counting of cells (ImageJ version 1.52; National Institutes of Health, Bethesda, MD). No a priori calculations were performed for sample size analysis. Normality was assessed by Shapiro-Wilk and Q-Q plots prior to parametric testing by Student’s t test or 1- or 2-way analysis of variance followed by post hoc testing as appropriate for the number of groups. All microfluidic qPCR data was analyzed using 2-way analysis of variance. The false discovery rate for the microfluidic qPCR was controlled with the 2-stage step-up method of Benjamini, Krieger, and Yekutieli.
All authors had access to the study data and reviewed and approved the final manuscript prior to submission.
CRediT Authorship Contributions
Breanna Sheahan, DVM, PhD (Conceptualization: Equal; Formal analysis: Lead; Investigation: Lead; Methodology: Lead; Visualization: Equal; Writing – original draft: Lead; Writing – review & editing: Lead)
Ally N Freeman (Formal analysis: Supporting; Investigation: Supporting; Validation: Supporting; Writing – review & editing: Supporting)
Theresa M Keeley (Investigation: Supporting; Writing – review & editing: Supporting)
Linda C Samuelson, PhD (Funding acquisition: Supporting; Resources: Supporting; Supervision: Supporting; Writing – review & editing: Supporting)
Jatin Roper, MD (Funding acquisition: Supporting; Resources: Supporting; Supervision: Supporting; Writing – review & editing: Supporting)
Stephanie Hasapis (Investigation: Supporting; Writing – review & editing: Supporting)
Chang-Lung Lee, PhD (Funding acquisition: Supporting; Resources: Supporting; Supervision: Supporting; Writing – review & editing: Supporting)
Christopher Matthew Dekaney, PhD (Conceptualization: Lead; Funding acquisition: Lead; Resources: Lead; Supervision: Lead; Writing – review & editing: Lead).
Article info
Publication history
Published online: February 08, 2021
Accepted:
January 19,
2021
Received:
June 16,
2020
Footnotes
Conflicts of Interest The authors disclose no conflicts.
Funding This work was supported by R01DK100508 from the National Institute of Diabetes and Digestive and Kidney Diseases (Christopher M. Dekaney), R01DK118023 from the National Institute of Diabetes and Digestive and Kidney Diseases (to Linda C. Samuelson), P30-DK34933 from the National Institute of Diabetes and Digestive and Kidney Diseases (to Linda C. Samuelson), and W81XWH-19-1-0170 from the Department of Defense (Chang-Lung Lee); the Whitehead Scholar Award from the Duke University School of Medicine (Jatin Roper); a pilot grant from the Opportunity Funds Management Core of the Centers for Medical Countermeasures against Radiation, National Institute of Allergy and Infectious Diseases (grant number U19AI067773 [to Chang-Lung Lee and Jatin Roper]); a Comparative Medicine and Translational Research Training Program fellowship (T32OD011130 [to Breanna J. Sheahan]); and an Undergraduate Summer Research Award through the North Carolina State University College of Veterinary Medicine (to Ally N. Freeman).
Copyright
© 2021 The Authors. Published by Elsevier Inc. on behalf of the AGA Institute.