Introduction
Many adult tissues harbor resident stem or progenitor cells that maintain tissue integrity and homeostasis (
1- Blanpain C.
- Horsley V.
- Fuchs E.
Epithelial stem cells: turning over new leaves.
). Whether the adult pancreas also harbors progenitor cells, however, is controversial, with evidence both for (
2- Sancho R.
- Gruber R.
- Gu G.
- Behrens A.
Loss of Fbw7 reprograms adult pancreatic ductal cells into α, δ, and β cells.
,
3- Xu X.
- D’Hoker J.
- Stangé G.
- Bonné S.
- De Leu N.
- Xiao X.
- Van de Casteele M.
- Mellitzer G.
- Ling Z.
- Pipeleers D.
- Bouwens L.
- Scharfmann R.
- Gradwohl G.
- Heimberg H.
Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas.
4- Criscimanna A.
- Speicher J.A.
- Houshmand G.
- Shiota C.
- Prasadan K.
- Ji B.
- Logsdon C.D.
- Gittes G.K.
- Esni F.
Duct cells contribute to regeneration of endocrine and acinar cells following pancreatic damage in adult mice.
) and against (
5- Kopp J.L.
- Dubois C.L.
- Schaffer A.E.
- Hao E.
- Shih H.P.
- Seymour P.A.
- Ma J.
- Sander M.
Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas.
,
6- Dor Y.
- Brown J.
- Martinez O.I.
- Melton D.A.
Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation.
7- Xiao X.
- Chen Z.
- Shiota C.
- Prasadan K.
- Guo P.
- El-Gohary Y.
- Paredes J.
- Welsh C.
- Wiersch J.
- Gittes G.K.
No evidence for beta cell neogenesis in murine adult pancreas.
) their existence
in vivo. Adult pancreatic progenitor cells could be a potential source of β-like cells for endogenous neogenesis or for cell replacement therapy of type 1 diabetes.
Although the
in vivo evidence is controversial, our laboratory has provided data that a rare population of cells in the adult murine pancreas has stem cell-like activities
in vitro: they self-renew long-term and give rise to cells that resemble all three major pancreatic lineages (
i.e. duct, acinar, and endocrine), including insulin-producing β-like cells (
8- Jin L.
- Feng T.
- Shih H.P.
- Zerda R.
- Luo A.
- Hsu J.
- Mahdavi A.
- Sander M.
- Tirrell D.A.
- Riggs A.D.
- Ku H.T.
Colony-forming cells in the adult mouse pancreas are expandable in Matrigel and form endocrine/acinar colonies in laminin hydrogel.
,
9- Jin L.
- Gao D.
- Feng T.
- Tremblay J.R.
- Ghazalli N.
- Luo A.
- Rawson J.
- Quijano J.C.
- Chai J.
- Wedeken L.
- Hsu J.
- LeBon J.
- Walker S.
- Shih H.P.
- Mahdavi A.
- et al.
Cells with surface expression of CD133(high)CD71(low) are enriched for tripotent colony-forming progenitor cells in the adult murine pancreas.
). We have named these rare tri-potent progenitors pancreatic colony-forming units (PCFUs),
3The abbreviations used are: PCFU
pancreatic colony-forming unit
PI3K
phosphoinositide 3-kinase
AKT
Ser/Thr kinase
DMEM
Dulbecco's modified Eagle's medium
FCS
fetal calf serum
qRT
quantitative PCR
EdU
5-ethynyl-2′-deoxyuridine
TUNEL
deoxynucleotidyltransferase-mediated dUTP nick end-labeling
FAK
focal adhesion kinase
DAPI
4′,6-diamidino-2-phenylindole
E/A
endocrine/acinar
IPA
Ingenuity Pathway Analysis
shβ-cats
shRNAs against β-catenin
NSC
neural stem cell
HSC
hematopoietic stem cell
hESC
human embryonic stem cell
RPKM
reads per kilobase per million
PE
phycoerythrin.
because they, as single cells, are able to form colonies (also known as organoids) in a methylcellulose-containing semisolid medium. Methylcellulose is a biologically-inert material that increases the viscosity of the medium. The semisolid medium prevents single cells from moving and aggregating, yet it is soft enough to allow a single cell to form a colony of cells in a three-dimensional space. Using the ability to study the
in vitro functions of adult murine PCFUs, this study aimed to investigate the molecular mechanism by which adult PCFUs self-renew.
GLIS family zinc finger 3 (Glis3) is a member of the Krüppel-like zinc finger transcription factor family (
10- Kim Y.S.
- Nakanishi G.
- Lewandoski M.
- Jetten A.M.
GLIS3, a novel member of the GLIS subfamily of Kruppel-like zinc finger proteins with repressor and activation functions.
). In postnatal mice, Glis3 plays an important role in the self-renewal of sperm stem cells (
11- Kang H.S.
- Chen L.Y.
- Lichti-Kaiser K.
- Liao G.
- Gerrish K.
- Bortner C.D.
- Yao H.H.
- Eddy E.M.
- Jetten A.M.
Transcription factor GLIS3: a new and critical regulator of postnatal stages of mouse spermatogenesis.
). Also, when expressed in conjunction with the “Yamanaka factors” Sox2, Klf4, c-Myc, and Oct4, Glis3 enhances the reprogramming of adult human adipose–derived stromal cells into induced pluripotent stem cells (
12- Lee S.Y.
- Noh H.B.
- Kim H.T.
- Lee K.I.
- Hwang D.Y.
Glis family proteins are differentially implicated in the cellular reprogramming of human somatic cells.
), suggesting an important role of Glis3 in the self-renewal and pluripotency of various stem/progenitor cells. In humans, Glis3 is important for proper endocrine cell development in the pancreas. Mutations in Glis3 are linked to neonatal diabetes (
13- Senée V.
- Chelala C.
- Duchatelet S.
- Feng D.
- Blanc H.
- Cossec J.C.
- Charon C.
- Nicolino M.
- Boileau P.
- Cavener D.R.
- Bougnères P.
- Taha D.
- Julier C.
Mutations in GLIS3 are responsible for a rare syndrome with neonatal diabetes mellitus and congenital hypothyroidism.
), a higher risk of type 2 diabetes in several genome-wide association studies (
14Emerging roles of GLIS3 in neonatal diabetes, type 1 and type 2 diabetes.
), beta-cell apoptosis (
15- Nogueira T.C.
- Paula F.M.
- Villate O.
- Colli M.L.
- Moura R.F.
- Cunha D.A.
- Marselli L.
- Marchetti P.
- Cnop M.
- Julier C.
- Eizirik D.L.
GLIS3, a susceptibility gene for type 1 and type 2 diabetes, modulates pancreatic beta cell apoptosis via regulation of a splice variant of the BH3-only protein Bim.
), and in some cases defects in the formation of both the exocrine and endocrine pancreas (
16- Dimitri P.
- Habeb A.M.
- Garbuz F.
- Millward A.
- Wallis S.
- Moussa K.
- Akcay T.
- Taha D.
- Hogue J.
- Slavotinek A.
- Wales J.K.
- Shetty A.
- Hawkes D.
- Hattersley A.T.
- Ellard S.
- De Franco E.
Expanding the clinical spectrum associated with GLIS3 mutations.
,
17- Dimitri P.
- Warner J.T.
- Minton J.A.
- Patch A.M.
- Ellard S.
- Hattersley A.T.
- Barr S.
- Hawkes D.
- Wales J.K.
- Gregory J.W.
Novel GLIS3 mutations demonstrate an extended multisystem phenotype.
). Glis3 is also necessary for proper pancreas development in mice, and mutations in Glis3 lead to decreased numbers of beta-cells, smaller islet volume, and subsequent neonatal diabetes (
18- Kang H.S.
- Kim Y.S.
- ZeRuth G.
- Beak J.Y.
- Gerrish K.
- Kilic G.
- Sosa-Pineda B.
- Jensen J.
- Pierreux C.E.
- Lemaigre F.P.
- Foley J.
- Jetten A.M.
Transcription factor Glis3, a novel critical player in the regulation of pancreatic beta-cell development and insulin gene expression.
,
19- Watanabe N.
- Hiramatsu K.
- Miyamoto R.
- Yasuda K.
- Suzuki N.
- Oshima N.
- Kiyonari H.
- Shiba D.
- Nishio S.
- Mochizuki T.
- Yokoyama T.
- Maruyama S.
- Matsuo S.
- Wakamatsu Y.
- Hashimoto H.
A murine model of neonatal diabetes mellitus in Glis3-deficient mice.
). Adult mice continually express Glis3 in the beta and duct cells of the pancreas (
20- Kang H.S.
- Takeda Y.
- Jeon K.
- Jetten A.M.
The spatiotemporal pattern of Glis3 expression indicates a regulatory function in bipotent and endocrine progenitors during early pancreatic development and in beta, PP and ductal cells.
). In both adult and embryonic mice, mutations or deletion of Glis3 leads to beta-cell apoptosis and a cystic duct phenotype (
18- Kang H.S.
- Kim Y.S.
- ZeRuth G.
- Beak J.Y.
- Gerrish K.
- Kilic G.
- Sosa-Pineda B.
- Jensen J.
- Pierreux C.E.
- Lemaigre F.P.
- Foley J.
- Jetten A.M.
Transcription factor Glis3, a novel critical player in the regulation of pancreatic beta-cell development and insulin gene expression.
,
19- Watanabe N.
- Hiramatsu K.
- Miyamoto R.
- Yasuda K.
- Suzuki N.
- Oshima N.
- Kiyonari H.
- Shiba D.
- Nishio S.
- Mochizuki T.
- Yokoyama T.
- Maruyama S.
- Matsuo S.
- Wakamatsu Y.
- Hashimoto H.
A murine model of neonatal diabetes mellitus in Glis3-deficient mice.
,
21- Kim Y.S.
- Kang H.S.
- Takeda Y.
- Hom L.
- Song H.Y.
- Jensen J.
- Jetten A.M.
Glis3 regulates neurogenin 3 expression in pancreatic beta-cells and interacts with its activator, Hnf6.
). Using global gene expression analysis in one of our prior studies, Glis3 was found to be expressed to a higher degree in the pancreatic CD133
highCD71
low ductal cell population, which is highly enriched for adult murine PCFUs (
9- Jin L.
- Gao D.
- Feng T.
- Tremblay J.R.
- Ghazalli N.
- Luo A.
- Rawson J.
- Quijano J.C.
- Chai J.
- Wedeken L.
- Hsu J.
- LeBon J.
- Walker S.
- Shih H.P.
- Mahdavi A.
- et al.
Cells with surface expression of CD133(high)CD71(low) are enriched for tripotent colony-forming progenitor cells in the adult murine pancreas.
), than in other populations. Given the pleiotropic roles of Glis3 in multiple biological processes and organs of different ages, we hypothesized that Glis3 was also important in the
in vitro self-renewal of adult murine PCFUs.
To test our hypothesis, lentiviral vectors carrying short-hairpin interfering RNAs (shRNAs) were used to knock down Glis3 expression, and the effects on adult murine PCFUs were measured. We found that Glis3 was required for the long-term self-renewal of PCFUs in vitro. Further analyses on the role of Glis3 in self-renewal revealed that Glis3 affected Wnt signaling molecules and, surprisingly, Prominin-1 (CD133). Follow-up studies using shRNAs against CD133 and Wnt effector β-catenin demonstrated a previously unknown Glis3–to–CD133–to-Wnt-signaling pathway that is required for maintaining the responsiveness to self-renewing signals of adult murine PCFUs.
Discussion
Using FACS-sorted PCFUs from adult murine pancreata and our unique colony assays, we demonstrate here that a Glis3–CD133–Wnt signaling axis regulates PCFU self-renewal. Glis3 (
11- Kang H.S.
- Chen L.Y.
- Lichti-Kaiser K.
- Liao G.
- Gerrish K.
- Bortner C.D.
- Yao H.H.
- Eddy E.M.
- Jetten A.M.
Transcription factor GLIS3: a new and critical regulator of postnatal stages of mouse spermatogenesis.
), CD133 (
43- Wei Y.
- Jiang Y.
- Zou F.
- Liu Y.
- Wang S.
- Xu N.
- Xu W.
- Cui C.
- Xing Y.
- Liu Y.
- Cao B.
- Liu C.
- Wu G.
- Ao H.
- Zhang X.
- Jiang J.
Activation of PI3K/Akt pathway by CD133-p85 interaction promotes tumorigenic capacity of glioma stem cells.
), and Wnt (
31- 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.
) have individually been implicated in regulating the self-renewal of stem and progenitor cells in tissues other than the pancreas. The current results, to the best of our knowledge, appear to be the first to delineate the sequence of signaling hierarchy among these molecules in regulating the self-renewal of progenitors. However, we caution that due to technical difficulties, we could not rescue Glis3 knockdown with overexpression of CD133; therefore, the specificity of the effect of Glis3 mediated through CD133 requires further investigation.
This study confirmed the necessity of canonical Wnt signaling for PCFU self-renewal
in vitro (
Fig. 5A). Previously, the increase of self-renewal by exogenous Wnt ligand (RSPO1) in adult murine PCFUs has been demonstrated in our published studies (
8- Jin L.
- Feng T.
- Shih H.P.
- Zerda R.
- Luo A.
- Hsu J.
- Mahdavi A.
- Sander M.
- Tirrell D.A.
- Riggs A.D.
- Ku H.T.
Colony-forming cells in the adult mouse pancreas are expandable in Matrigel and form endocrine/acinar colonies in laminin hydrogel.
), and the same conclusion was corroborated by other studies (
56- Huch M.
- Bonfanti P.
- Boj S.F.
- Sato T.
- Loomans C.J.
- van de Wetering M.
- Sojoodi M.
- Li V.S.
- Schuijers J.
- Gracanin A.
- Ringnalda F.
- Begthel H.
- Hamer K.
- Mulder J.
- van Es J.H.
- et al.
Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis.
,
57- Dorrell C.
- Tarlow B.
- Wang Y.
- Canaday P.S.
- Haft A.
- Schug J.
- Streeter P.R.
- Finegold M.J.
- Shenje L.T.
- Kaestner K.H.
- Grompe M.
The organoid-initiating cells in mouse pancreas and liver are phenotypically and functionally similar.
). In addition to the Wnt findings, the new findings from this study are the roles of Glis3 and CD133 for
in vitro self-renewal of adult murine PCFUs.
Our data add to a growing body of literature demonstrating the importance of Glis3 in the self-renewal and maintenance of progenitor cells. As mentioned, a mutation of Glis3 in murine sperm stem cells results in a decreased expression of genes important for self-renewal (
i.e. Lhx1,
Sall4, and
Id4), leading to a severe lack of mature sperm cells (
11- Kang H.S.
- Chen L.Y.
- Lichti-Kaiser K.
- Liao G.
- Gerrish K.
- Bortner C.D.
- Yao H.H.
- Eddy E.M.
- Jetten A.M.
Transcription factor GLIS3: a new and critical regulator of postnatal stages of mouse spermatogenesis.
). Overexpression of Glis3 increases reprogramming efficiency of canonical Yamanaka factors (
12- Lee S.Y.
- Noh H.B.
- Kim H.T.
- Lee K.I.
- Hwang D.Y.
Glis family proteins are differentially implicated in the cellular reprogramming of human somatic cells.
), presumably through the up-regulation of FoxA2, Wnt, and the induction of pluripotency genes, similar to those induced by the family member Glis1 (
58- Maekawa M.
- Yamaguchi K.
- Nakamura T.
- Shibukawa R.
- Kodanaka I.
- Ichisaka T.
- Kawamura Y.
- Mochizuki H.
- Goshima N.
- Yamanaka S.
Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1.
), which has 94% homology to Glis3 (
10- Kim Y.S.
- Nakanishi G.
- Lewandoski M.
- Jetten A.M.
GLIS3, a novel member of the GLIS subfamily of Kruppel-like zinc finger proteins with repressor and activation functions.
,
59- Kim Y.S.
- Lewandoski M.
- Perantoni A.O.
- Kurebayashi S.
- Nakanishi G.
- Jetten A.M.
Identification of Glis1, a novel Gli-related, Kruppel-like zinc finger protein containing transactivation and repressor functions.
).
Here, we show that Glis3 regulates self-renewal of adult murine PCFUs in organoids by changing the gene and protein expression of certain Wnt genes (
Figure 1,
Figure 4). Interestingly, the negative effect of Glis3 knockdown on self-renewal occurred later at the 4th generation, compared with CD133 and β-catenin knockdowns at the 2nd generation. Knockdown of Glis3 down-regulates and up-regulates Wnt-related genes in the 1st and 4th generations, respectively. These contrasting results are likely due to the known pleiotropic effects of Glis3 impacting multiple pathways (
60GLIS1–3 transcription factors: critical roles in the regulation of multiple physiological processes and diseases.
,
61- Rurale G.
- Persani L.
- Marelli F.
GLIS3 and thyroid: a pleiotropic candidate gene for congenital hypothyroidism.
). We speculate that Glis3 fine-tunes its effects over time in a cell-dependent manner: in the earlier PCFUs to sustain the expression of Wnt receptors (
e.g. Lgr5 and Lrp6) as well as CD133, and, once the PCFUs are exhausted or in the differentiated ductal cells, inhibiting proliferation via a currently unknown mechanism.
While our manuscript was in revision, Jeon
et al. (
62- Jeon K.
- Kumar D.
- Conway A.E.
- Park K.
- Jothi R.
- Jetten A.M.
GLIS3 transcriptionally activates WNT genes to promote differentiation of human embryonic stem cells into posterior neural progenitors.
) showed that overexpression of Glis3 in human embryonic stem cells (hESCs) causes up-regulation of Wnt ligand genes, such as
Wnt3A. This increase was caused by a direct binding of Glis3 to the Wnt3A promoter, demonstrated by ChIP-seq analysis using FLAG-tagged Glis3.
In silico analysis utilizing JASPAR motifs and the eukaryotic promoter database demonstrated potential Glis3 consensus-binding sites in promoter regions of
CD133,
Axin2,
Lgr5, and β-catenin (data not shown), genes not reported by Jeon
et al. (
62- Jeon K.
- Kumar D.
- Conway A.E.
- Park K.
- Jothi R.
- Jetten A.M.
GLIS3 transcriptionally activates WNT genes to promote differentiation of human embryonic stem cells into posterior neural progenitors.
). Again, it is possible that the effects of Glis3 are dependent on cellular context, and further analysis will be necessary to compare how Glis3 may differentially affect Wnt genes in our PCFUs
versus hESCs.
The role of CD133 in self-renewal and proliferation has been well-documented in tumor-initiating cells (
63- Brescia P.
- Ortensi B.
- Fornasari L.
- Levi D.
- Broggi G.
- Pelicci G.
CD133 is essential for glioblastoma stem cell maintenance.
). Overexpression of CD133 in carcinoma cell lines originating from the mouth, head, and neck increases the expression of stem cell-specific genes,
Oct4,
Sox2, and
Nanog (
64- Moon Y.
- Kim D.
- Sohn H.
- Lim W.
Effect of CD133 overexpression on the epithelial–to-mesenchymal transition in oral cancer cell lines.
,
65- Lee J.
- Park M.
- Ko Y.
- Kim B.
- Kim O.
- Hyun H.
- Kim D.
- Sohn H.
- Moon Y.L.
- Lim W.
Ectopic overexpression of CD133 in HNSCC makes it resistant to commonly used chemotherapeutics.
). CD133 has also been shown to be necessary for the proliferation and self-renewal of various tumor cells and human embryonic kidney cells through interacting with HDAC6 to influence Wnt signaling (
41- Mak A.B.
- Nixon A.M.
- Kittanakom S.
- Stewart J.M.
- Chen G.I.
- Curak J.
- Gingras A.C.
- Mazitschek R.
- Neel B.G.
- Stagljar I.
- Moffat J.
Regulation of CD133 by HDAC6 promotes β-catenin signaling to suppress cancer cell differentiation.
). Finally, knockdown of CD133 in glioblastoma tumor cells caused a significant decrease in self-renewal (
63- Brescia P.
- Ortensi B.
- Fornasari L.
- Levi D.
- Broggi G.
- Pelicci G.
CD133 is essential for glioblastoma stem cell maintenance.
) demonstrating that in tumor cells CD133 is required for stemness properties.
Despite the wealth of the above-mentioned research in tumor cells, the functional role of CD133 has not been extensively explored in normal murine tissues. In the mouse embryo, CD133 affects neural stem cell (NSC) maintenance through changes to asymmetrical division where NSCs retain and differentiated young neurons lack CD133 (
66- Kosodo Y.
- Röper K.
- Haubensak W.
- Marzesco A.M.
- Corbeil D.
- Huttner W.B.
Asymmetric distribution of the apical plasma membrane during neurogenic divisions of mammalian neuroepithelial cells.
). This may suggest a positive role for CD133 in self-renewal of embryonic NSCs. In adult hematopoietic stem cells (HSCs), however, CD133 knockdown does not affect the self-renewal of HSCs but leads to an enhanced differentiation of HSCs toward the myeloid, as opposed to the lymphoid, lineage (
67- Arndt K.
- Grinenko T.
- Mende N.
- Reichert D.
- Portz M.
- Ripich T.
- Carmeliet P.
- Corbeil D.
- Waskow C.
CD133 is a modifier of hematopoietic progenitor frequencies but is dispensable for the maintenance of mouse hematopoietic stem cells.
). These studies, together with our current results (discussed more below), suggest that the roles of CD133 are likely cell type–specific.
In contrast to hESCs (
68- ten Berge D.
- Kurek D.
- Blauwkamp T.
- Koole W.
- Maas A.
- Eroglu E.
- Siu R.K.
- Nusse R.
Embryonic stem cells require Wnt proteins to prevent differentiation to epiblast stem cells.
), the colonies grown in our standard Matrigel assay (without exogenous RSPO1) do not appear to secrete endogenous Wnt ligands (
Fig. 1D), which should prevent a positive feed-forward loop. Importantly, the ratio of phosphorylated Lrp6 to total Lrp6 protein was not changed by either Glis3 or CD133 knockdown, suggesting that Wnt signaling initiation may be triggered without Glis3 or CD133. Additionally, we found that CD133 was required for optimal Lgr5 expression (
Figs. 2,
A and
B, and
6A), as well as maintaining some active form of β-catenin (
Fig. 6B), which is known to activate Lgr5 transcription (
31- 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.
). Glis3 maintained the expression of Lrp6 in addition to Lgr5 (
Fig. 1,
E and
F), highlighting overlapping yet divergent effects of Glis3 and CD133 in maintaining the expression of Wnt receptors. Because RSPO1 depends on Lgr5 and Lrp6 for signal transmission (
49- Pinson K.I.
- Brennan J.
- Monkley S.
- Avery B.J.
- Skarnes W.C.
An LDL-receptor-related protein mediates Wnt signalling in mice.
), these results suggest that Glis3 and CD133 collectively endow PCFUs the ability to respond to external Wnt stimuli by maintaining the expression of these Wnt receptors.
To the best of our knowledge, our study is the first to demonstrate the physical association of CD133 with β-catenin via E-cadherin in primary murine cells. This association may suggest functional interactions between CD133 and β-catenin, although we could not detect a direct physical interaction between these two molecules by co-immunoprecipitation analysis (data not shown). Previously, it has been shown that E-cadherin and β-catenin interact at adherens junctions on the cell membrane to increase cell–cell adhesion (
54Convergence of Wnt, β-catenin, and cadherin pathways.
). When bound to E-cadherin, β-catenin is typically considered “sequestered” and not able to stimulate Wnt signaling (
69- Tang Y.
- Liu Z.
- Zhao L.
- Clemens T.L.
- Cao X.
Smad7 stabilizes β-catenin binding to E-cadherin complex and promotes cell-cell adhesion.
). Knockdown of CD133 did not affect E-cadherin levels in our cells (
Fig. 6B), suggesting that the sequestering mechanism of β-catenin by E-cadherin is in place. However, a switch in β-catenin function from adhesion to Wnt signaling may be regulated by phosphorylating β-catenin at specific tyrosine residues (
70- Brembeck F.H.
- Schwarz-Romond T.
- Bakkers J.
- Wilhelm S.
- Hammerschmidt M.
- Birchmeier W.
Essential role of BCL9–2 in the switch between β-catenin’s adhesive and transcriptional functions.
,
71- Taddei M.L.
- Chiarugi P.
- Cirri P.
- Buricchi F.
- Fiaschi T.
- Giannoni E.
- Talini D.
- Cozzi G.
- Formigli L.
- Raugei G.
- Ramponi G.
β-Catenin interacts with low-molecular-weight protein tyrosine phosphatase leading to cadherin-mediated cell-cell adhesion increase.
). CD133 can act as a scaffold for other serine/threonine kinases, including AKT (
43- Wei Y.
- Jiang Y.
- Zou F.
- Liu Y.
- Wang S.
- Xu N.
- Xu W.
- Cui C.
- Xing Y.
- Liu Y.
- Cao B.
- Liu C.
- Wu G.
- Ao H.
- Zhang X.
- Jiang J.
Activation of PI3K/Akt pathway by CD133-p85 interaction promotes tumorigenic capacity of glioma stem cells.
), which has been shown to directly activate β-catenin (
43- Wei Y.
- Jiang Y.
- Zou F.
- Liu Y.
- Wang S.
- Xu N.
- Xu W.
- Cui C.
- Xing Y.
- Liu Y.
- Cao B.
- Liu C.
- Wu G.
- Ao H.
- Zhang X.
- Jiang J.
Activation of PI3K/Akt pathway by CD133-p85 interaction promotes tumorigenic capacity of glioma stem cells.
,
72- Fang D.
- Hawke D.
- Zheng Y.
- Xia Y.
- Meisenhelder J.
- Nika H.
- Mills G.B.
- Kobayashi R.
- Hunter T.
- Lu Z.
Phosphorylation of β-catenin by AKT promotes β-catenin transcriptional activity.
). Additionally, CD133 can directly interact with the tyrosine kinase Src
in vitro (
73- Liu C.
- Li Y.
- Xing Y.
- Cao B.
- Yang F.
- Yang T.
- Ai Z.
- Wei Y.
- Jiang J.
The interaction between cancer stem cell marker CD133 and Src protein promotes focal adhesion kinase (FAK) phosphorylation and cell migration.
), which has been shown to promote activation of focal adhesion kinase (FAK), leading to β-catenin nuclear localization and murine epidermal cell migration (
74- Ridgway R.A.
- Serrels B.
- Mason S.
- Kinnaird A.
- Muir M.
- Patel H.
- Muller W.J.
- Sansom O.J.
- Brunton V.G.
Focal adhesion kinase is required for β-catenin-induced mobilization of epidermal stem cells.
). We therefore speculate that certain tyrosine kinases associated with CD133 can play a direct role in activating β-catenin.
Alternatively, CD133-mediated β-catenin activation may be controlled through GSK-3β inhibition. All activities of GSK-3β, including that in the destruction complex for β-catenin, are inhibited by CHIR99021 (
53- Ring D.B.
- Johnson K.W.
- Henriksen E.J.
- Nuss J.M.
- Goff D.
- Kinnick T.R.
- Ma S.T.
- Reeder J.W.
- Samuels I.
- Slabiak T.
- Wagman A.S.
- Hammond M.E.
- Harrison S.D.
Selective glycogen synthase kinase 3 inhibitors potentiate insulin activation of glucose transport and utilization in vitro and in vivo.
,
75- Chen E.Y.
- DeRan M.T.
- Ignatius M.S.
- Grandinetti K.B.
- Clagg R.
- McCarthy K.M.
- Lobbardi R.M.
- Brockmann J.
- Keller C.
- Wu X.
- Langenau D.M.
Glycogen synthase kinase 3 inhibitors induce the canonical WNT/β-catenin pathway to suppress growth and self-renewal in embryonal rhabdomyosarcoma.
). The observation that CHIR99021, when added to colonies with CD133 knockdown, rescued Wnt target gene expression (
Fig. 6A) suggests that CD133 may inhibit GSK-3β, but this hypothesis requires further investigation.
In this study CD133, but not Glis3 or β-catenin, is required for both self-renewal (
Fig. 3,
A and
C) and PI3K/AKT-mediated survival (
Fig. 3,
B,
D, and
E), demonstrating the functional importance of CD133 for adult murine PCFUs. However, the fact that only cells that express high levels of CD133 on their cell surface can form colonies, but not every single CD133
high cell is capable of forming a colony (
Fig. S1, A and B) (
26- Jin L.
- Feng T.
- Zerda R.
- Chen C.C.
- Riggs A.D.
- Ku H.T.
In vitro multilineage differentiation and self-renewal of single pancreatic colony-forming cells from adult C57Bl/6 mice.
), suggests that CD133 is necessary but not sufficient to confer a PCFU status. Whether Glis3 is a molecule that enables CD133
high cells to form colonies needs further investigation.
IPA analyses of Glis3- and CD133-affected genes in 3-week-old colonies grown in Matrigel and RSPO1 revealed interesting biological pathways and upstream regulators (
Figs. 1,
G and
H, and
2,
C and
D). Cell movement, migration, and NF-κB, a key regulator in immune response (
76The nuclear factor NF-κB pathway in inflammation.
), are affected by the knockdowns of both Glis3 and CD133, which may implicate the involvement in these processes by their common downstream signal, Wnt. These biological processes are not typically associated with stemness of other organs. However, prior findings did show roles of Wnt in cytoskeletal rearrangement and chemokine expression, which directly affected cell movement and migration in various cell types (
77- Sedgwick A.E.
- D'Souza-Schorey C.
Wnt signaling in cell motility and invasion: drawing parallels between development and cancer.
), as well as cross-talk in the inflammatory responses (
78Crosstalk between Wnt/β-catenin and NF-κB signaling pathway during inflammation.
). The upstream complex analyses in the CD133-knockdown genes identified the possible involvement of both PI3K/AKT and Stat3 signaling (
Fig. S4). However, only the activation of AKT but not Stat3 was affected by CD133 knockdown in colonies grown in Matrigel (
Fig. 2B). This result suggests that, although dependent on CD133, AKT signaling affects downstream genes independent of Stat3. It is known that Stat3 can directly affect AKT expression (
79STAT3 target genes relevant to human cancers.
), and therefore, the IPA analysis may have indiscriminately identified Stat3 by association. Regardless, the other pathways identified in the IPA analyses will open new avenues to further decipher the biology of adult PCFUs.
In summary, this study reveals a Glis3–CD133–Wnt molecular pathway and its functions in
in vitro self-renewal and survival of adult murine PCFUs (
Fig. 7). These results may have implications for targeting this signaling axis for maintenance and expansion of adult PCFUs for endogenous neogenesis or cell-replacement therapy of type 1 diabetes, in which insulin-producing beta cells are destroyed by an autoimmune attack (
80The pathogenesis and natural history of type 1 diabetes.
).
Experimental procedures
Mice
C57BL/6J mice (The Jackson Laboratory, Bar Harbor, ME) aged 8–12 weeks old from both sexes were used in this study. All mice were maintained under specific pathogen-free conditions, and animal experiments were conducted according to the Institutional Animal Care and Use Committee at the City of Hope. All procedures and protocols were approved by the Institutional Animal Care and Use Committee at the City of Hope.
Dissociation of pancreas into single cells
Murine pancreata were dissected, cleared of fat tissue under a dissecting microscope, and rinsed three times in cold Dulbecco’s PBS (DPBS) containing 0.1% bovine serum albumin (BSA), 100 units/ml penicillin, and 100 μg/ml streptomycin; this wash solution is referred to as PBS/BSA. Pancreata were minced in a dry Petri dish placed on ice using spring scissors for 3 min or until finely minced. The tissue was transferred to a 50-ml conical tube and resuspended in PBS/BSA containing collagenase B (2–4 mg/ml) (Roche Applied Science, Mannheim, Germany) and DNase I (2000 units/ml) (Calbiochem, Darmstadt, Germany). Tissue was incubated at 37 °C for 16 min with swirling every 2–3 min and gently passed through a 16-gauge syringe needle every 8 min. Cells were then washed in cold PBS/BSA supplemented with 2000 units/ml DNase I and successively passed through a 100- and a 40-μm mesh filter (BD Biosciences) to yield a mostly single cell suspension.
Cell sorting
Sorting was performed similarly to our previous publication (
9- Jin L.
- Gao D.
- Feng T.
- Tremblay J.R.
- Ghazalli N.
- Luo A.
- Rawson J.
- Quijano J.C.
- Chai J.
- Wedeken L.
- Hsu J.
- LeBon J.
- Walker S.
- Shih H.P.
- Mahdavi A.
- et al.
Cells with surface expression of CD133(high)CD71(low) are enriched for tripotent colony-forming progenitor cells in the adult murine pancreas.
). Briefly, dissociated pancreatic cells were first incubated with anti-mouse CD16/32 (10 μg/ml) (BioLegend, San Diego) for 5 min on ice to diminish nonspecific binding. Biotin-conjugated anti-mouse CD133 (clone 13A4; 5 μg/ml; eBioscience, San Diego) and phycoerythrin/Cy7 (PECy7)-conjugated anti-mouse CD71 (clone RI7217; 5 μg/ml; BioLegend, San Diego) antibodies were added. Cells were incubated for 20 min on ice, washed twice, then treated with streptavidin-labeled allophycocyanin (2 μg/ml BioLegend, San Diego) for 15 min on ice, washed twice and resuspended in PBS/BSA/DNase I containing DAPI (0.2 μg/ml). Control antibodies were biotin-conjugated rat IgG1 (5 μg/ml; eBioscience, San Diego) and PE/Cy7-conjugated rat IgG1 (5 μg/ml; BioLegend, San Diego). Acquired flow cytometry data were analyzed with software provided by FlowJo (TreeStar, Ashland, OR). Cell sorting was performed on an Aria special-order research product (BD Biosciences). All analyses included an initial gating of forward and side scatters to exclude debris. In cell-sorting experiments, doublets were further excluded by gating out high pulse-width cells, and live cells were selected by DAPI-negative staining (
Fig. S5).
Colony assay
Sorted cells were resuspended at a density of 2.5 × 10
3 cells/well/0.5 ml for the Matrigel assay or 2.5 × 10
4 cells/well/0.5 ml for the laminin-hydrogel assay as described previously (
8- Jin L.
- Feng T.
- Shih H.P.
- Zerda R.
- Luo A.
- Hsu J.
- Mahdavi A.
- Sander M.
- Tirrell D.A.
- Riggs A.D.
- Ku H.T.
Colony-forming cells in the adult mouse pancreas are expandable in Matrigel and form endocrine/acinar colonies in laminin hydrogel.
). Culture media contained DMEM/F-12 media, 1% methylcellulose (Sinetsu Chemical, Tokyo, Japan), 50% conditioned media from mouse embryonic stem cell-derived pancreatic-like cells (
81- Winkler M.
- Trieu N.
- Feng T.
- Jin L.
- Walker S.
- Singh L.
- Ku H.T.
A quantitative assay for insulin-expressing colony-forming progenitors.
), 5% fetal calf serum (FCS), 10 mmol/liter nicotinamide (Sigma), 10 ng/ml human recombinant activin B (R&D Systems, Minneapolis, MN), 0.1 nmol/liter exendin-4 (Sigma), 1 ng/ml vascular endothelial growth factor-A (Sigma), and 750 ng/ml R-spondin-1 (R&D Systems, Minneapolis, MN). When indicated, either 5% (v/v) Matrigel (referred to as the standard assay in this study) or 100 μg/ml laminin hydrogel (
8- Jin L.
- Feng T.
- Shih H.P.
- Zerda R.
- Luo A.
- Hsu J.
- Mahdavi A.
- Sander M.
- Tirrell D.A.
- Riggs A.D.
- Ku H.T.
Colony-forming cells in the adult mouse pancreas are expandable in Matrigel and form endocrine/acinar colonies in laminin hydrogel.
) was added to the media for the generation of cystic or endocrine/acinar colonies, respectively. Cells were plated in 24-well ultra-low protein-binding plates (Corning, New York) and incubated in a humidified 5% CO
2 atmosphere at 37 °C. Colonies grown in Matrigel or laminin hydrogel were counted 3 weeks or 10 days after plating, respectively.
Colony dissociation and replating
Warmed PBS/BSA (1 ml per well) was added to each well of a 24-well plate containing colonies. Cystic colonies were collected in a 50-ml conical tube, washed, resuspended in 10 ml of 2–4 mg/ml collagenase B, incubated for 15 min at 37 °C with mixing every 5 min, and washed in PBS/BSA. Subsequently, colonies were treated with 20 ml of 0.25% (w/v) trypsin-EDTA, incubated for 3 min at 37 °C, and pipetted thoroughly to generate mostly single cells. Warmed FCS (4 ml) was added to the cells to stop the trypsin digestion. Cells were washed in PBS/BSA and kept at room temperature. A portion was mixed with 0.02% (w/v) trypan blue, and the concentration of live cells (i.e. the trypan blue–negative cells) was determined by a hemocytometer. For replating experiments, the final single cell suspension was mixed with medium containing Matrigel or laminin hydrogel as indicated.
Lentivirus transduction
Lentiviral particles containing shRNAs against control (catalog no. SHC002V), Glis3 (SHCLNV-NM_175459), Prominin-1 (CD133) (SHCLNV-NM_008935), or CTNNB1 (β-catenin) (SHCLNV-NM_007614) were purchased (Sigma). Five independent clones of shRNAs were pooled with equal ratios. Freshly-sorted cells were seeded at a concentration of 100,000 cells/96-well plate/100 μl in a liquid medium (i.e. colony assay medium minus Matrigel, laminin hydrogel, RSPO1, and methylcellulose) in a flat-bottom low-attachment plate (Thermo Fisher Scientific, Waltham, MA) and incubated at 37 °C for 4 h. Viral particles were added at a mode of infection of 20. The final volumes were brought to 300 μl/well using DMEM/F-12 containing penicillin/streptomycin and 4 μg/ml hexadimethrine bromide (polybrene) (Sigma). Cells were incubated overnight at 37 °C, washed twice with warmed PBS/BSA, counted, and plated into a colony assay medium. Puromycin (2 μg/ml) was added to each well to select for infected cells.
Conventional or microfluidic quantitative (q)RT-PCR
For conventional and microfluidic qRT-PCR analyses, the same procedures were employed as reported previously (
8- Jin L.
- Feng T.
- Shih H.P.
- Zerda R.
- Luo A.
- Hsu J.
- Mahdavi A.
- Sander M.
- Tirrell D.A.
- Riggs A.D.
- Ku H.T.
Colony-forming cells in the adult mouse pancreas are expandable in Matrigel and form endocrine/acinar colonies in laminin hydrogel.
). Duplicate samples were used in all analyses. Microfluidic qRT-PCR was performed using the BioMark
TM 48.48 Dynamic Array system (Fluidigm, South San Francisco, CA). Single colonies were lifted one by one from the methylcellulose-containing medium under direct microscopic visualization by using a 10-μl Eppendorf pipette, collected in reaction buffer (10 μl), and followed by preamplification (12 cycles for Matrigel-grown cystic colonies and 18 cycles for E/A colonies) according to the manufacturer’s instructions (Fluidigm). Amplified cDNA was loaded onto a 48.48 Dynamic Array system using the NanoFlex integrated fluidic circuit controller (Fluidigm). Threshold cycle (
Ct), as a measure of fluorescence intensity, was determined by the BioMark PCR analysis software (Fluidigm) and expressed as Δ
Ct. All experiments were performed with negative (water) and positive (adult C57BL/6J pancreatic cells) controls. TaqMan probes used in this study are listed in
Table S1.
Western blotting
Cells were lysed on ice in radioimmunoprecipitation assay (RIPA) buffer (Thermo Fisher Scientific) containing 50 m
m Tris, pH 7.4, 150 m
m NaCl, 0.1 m
m EDTA, 1% Triton X-100, 0.5% sodium deoxycholate, 0.1% SDS, phosphatase inhibitor mixture III (Calbiochem/Merck, Bad Soden, Germany), and complete protease inhibitor (Roche Applied Science, Mannheim, Germany). Extracts were sedimented at 10,000 ×
g for 15 min at 4 °C to remove insoluble material. Protein concentrations were determined using the Pierce® BCA protein assay (Thermo Fisher Scientific). Samples of proteins (10 μg) were boiled in 1× sample buffer, separated by SDS-PAGE on a 10% Laemmli gel, transferred to a polyvinylidene difluoride membrane (Bio-Rad), incubated with primary and secondary antibodies, and developed using enhanced luminol chemiluminescence (ECL-kit; Bio-Rad). Images were detected using Crystal Blue X-ray film (Bio-Rad). Antibodies used in Western blotting are listed in
Table S2.
Co-immunoprecipitation
Three-week-old colonies were hand-picked, pooled, and lysed on ice in Pierce® IP lysis buffer (Thermo Fisher Scientific) containing phosphatase inhibitor mixture III (Calbiochem/Merck, Bad Soden, Germany) and complete protease inhibitor (Roche Applied Science, Mannheim, Germany) for 30 min with gentle pipetting every 10 min. Protein concentration was measured using the Pierce® BCA protein assay (Thermo Fisher Scientific). Lysates were pre-cleared using protein G magnetic beads (Cell Signaling Technology, Danvers, MA) for 1 h at 4 °C with rotation. The beads were pelleted via magnetic separation rack and the lysate was removed. Primary antibodies were added to the pre-cleared lysates overnight with rotation at 4 °C. The next day, protein G magnetic beads were added for 1 h at 4 °C with rotation, and the beads were pelleted using a magnetic separation rack. Beads containing pulled down proteins were then washed five times with lysis buffer and boiled at 95 °C for 15 min in 3× sample buffer. Samples were brought to 1× sample buffer and then run on SDS-PAGE gel for subsequent Western blot analysis.
Proliferation and apoptosis analyses
For proliferation assay, Click-iT EdU (Thermo Fisher Scientific) was added to cells grown in laminin hydrogel-containing colony assay, and 10 days after plating, individual E/A colonies were hand-picked, pooled, and fixed with 4% paraformaldehyde for 20 min at room temperature. Colonies were washed and stored at 4 °C in PBS containing 0.01% Triton X-100. The Alexa Fluor 594 imagine kit (Thermo Fisher Scientific) was used to visualize EdU+ cells. For apoptosis assay, fixed 10-day-old E/A colonies were stained using the Click-iT TUNEL Alexa Fluor 488 imagine kit (Thermo Fisher Scientific) according to the manufacturer’s instructions. For imaging, E/A colonies were resuspended in Hoechst solution, placed in 35-mm glass-bottom dishes, and serially imaged using the z-stack function on a Zeiss 700 LSM. Individual nuclei positive for Hoechst were analyzed for the presence of EdU or TUNEL, and the fraction of positive nuclei was recorded.
mRNA sequencing
Each biological sample submitted for RNA-Seq was collected from a total of five pancreata that had previously been dissociated into single cell suspension, sorted for CD133highCD71low cells, infected with lentivirus carrying either shControl, shGlis3s, or shCD133s, and plated into a standard Matrigel-containing colony assay in the presence of exogenous RSPO1. The resulting 3-week-old cystic colonies were pooled and prepared for sequencing. Two biological replicates were used from each knockdown. cDNA libraries were prepared with Kapa Stranded mRNA-seq kit (KAPA Biosystems, Wilmington, MA) according to the manufacturer’s protocol with minor modifications. Briefly, polyadenylated RNAs were enriched from 500 ng of total RNAs using oligo(dT) magnetic beads, fragmented with divalent cations under elevated temperature, and reverse-transcribed into the first-strand followed by the second-strand cDNAs. Subsequently, the double-stranded cDNAs underwent end repair and 3′-end adenylation, ligated with barcoded adaptors, and amplified by PCR (12 cycles). The cDNA libraries were validated by the Agilent Bioanalyzer (Agilent) and prepared for sequencing using cBot cluster generation system (Illumina) with HiSeq SR Cluster Version 4 kit (Illumina). Using the HiSeq 2500 platform with HiSeq SBS Version 4 kits (Illumina), the sequencing run was performed in the single read mode with 51 cycles of “read1” and seven cycles of “index read.” Real-time analysis 2.2.38 software (Illumina) was used to process the image analysis and base calling.
Data analysis for mRNA sequencing
Raw sequence reads were mapped to the mouse genome (mm10) using TopHat (
82- Trapnell C.
- Pachter L.
- Salzberg S.L.
TopHat: discovering splice junctions with RNA-Seq.
), and the frequency of Refseq genes was counted using HTseq. The raw counts were then normalized using the method of trimmed mean of M values (TMM) and compared using Bioconductor package “edgeR”. Reads per kilobase per million (RPKM) were also calculated from the raw counts. Differentially expressed genes were identified if the RPKM were ≥1 in at least one sample, fold-change of ≥2, and
p ≤ 0.05. These differential genes were then imported into Ingenuity Pathway Analysis for functional pathway analysis. Raw RNA-Seq results were deposited on the Gene Expression Omnibus (GEO) database (
https://www.ncbi.nlm.nih.gov/geo/) under the ID code GSE124944.
Statistical analysis
All values shown are represented as either mean ± S.D. or mean ± S.E. p values were calculated using Student’s two-tailed t test; p < 0.05 was considered significant.