A microRNA-operated switch of asymmetric-to-symmetric cancer stem cell divisions

3
NEWS AND VIEWS a widely used diabetic mouse model in which β-cells are not able to compensate for the mas- sive need for insulin (BKS.db/db mice), islet SIK2 protein levels were actually reduced in spite of overt diabetes (and thus hyperglycae- mia), indicating that SIK2 stabilization (and potentially activity) must be controlled by alternative mechanisms — not only by glu- cose levels. Thus, finding upstream regulators of SIK2 activity will be important, as it might also reveal molecular mechanisms leading to β-cell failure in insulin-resistant or diabetic states. It remains to be tested if forced expres- sion or stabilization of SIK2 in β-cells could ameliorate the diabetic phenotype of the BKS. db/db mouse or other models of obesity that cannot compensate for their insulin resist- ance. The work by Screaton and colleagues further suggests that PJA2 plays a major role in β-cell physiology. PJA2 activity is known to be modulated by intracellular cyclic adenosine monophosphate (cAMP) levels in vitro 14 , and cAMP levels in β-cells are actively regulated — for example, by the intestinal hormone GLP1 (glucagon-like peptide 1), whose long-lasting analogues are used as T2D therapeutics 15 . Whether SIK2 and PJA2 are co-expressed in human β-cells, and whether glucose-mediated stabilization of SIK2 occurs in this setting as well, remains to be tested experimentally. Furthermore, it will be interesting to examine if SIK2 also controls the secretion of other hor- mones regulating glucose homeostasis, includ- ing GLP1 secretion by intestinal L-cells. This study of SIK2’s function in β-cells clearly advances our knowledge of β-cell physiology, and future investigations into the regulation of SIK2 function may be fruitful in the hunt for new therapeutic approaches for treatment of T2D. COMPETING FINANCIAL INTERESTS The authors declare no competing financial interests. 1. Konner, A. C. & Bruning, J. C. Cell Metab. 16, 144–152 (2012). 2. Ashcroft, F. M. & Rorsman, P. Cell 148, 1160–1171 (2012). 3. Sakamaki, J. et al. Nat. Cell Biol. 16, 234–244 (2014). 4. Rathmann, W. & Giani, G. Diabetes Care 27, 2568– 2569 (2004). 5. Lumeng, C. N. & Saltiel, A. R. J. Clin. Invest. 121, 2111–2117 (2011). 6. Bright, N. J., Thornton, C. & Carling, D. Acta Physiol. 196, 15–26 (2009). 7. Ruderman, N. B., Carling, D., Prentki, M. & Cacicedo, J. M. J. Clin. Invest. 123, 2764–2772 (2013). 8. Sasaki, T. et al. Neuron 69, 106–119 (2011). 9. Bricambert, J. et al. J. Clin. Invest. 120, 4316–4331 (2010). 10. Horike, N. et al. J. Biol. Chem. 278, 18440–18447 (2003). 11. Kulkarni, R. N. et al. Cell 96, 329–339 (1999). 12. Okada, T. et al. Proc. Natl Acad. Sci. USA 104, 8977– 8982 (2007). 13. Wei, F. Y. et al. Nat. Med. 11, 1104–1108 (2005). 14. Lignitto, L. et al. Nat. Cell Biol. 13, 412–422 (2011). 15. Seino, S. Diabetologia 55, 2096–2108 (2012). Robin G. Lerner and Claudia Petritsch are in the Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Comprehensive Cancer Center, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California, USA. e-mail: [email protected] A microRNA-operated switch of asymmetric-to-symmetric cancer stem cell divisions Robin G. Lerner and Claudia Petritsch Defective asymmetric cell divisions of stem and progenitor cells are associated with tumorigenesis by a largely unknown mechanism. A signalling axis involving Snail, microRNA-146a and Numb is now shown to regulate the switch between symmetric and asymmetric cell division in colorectal cancer stem cells. Somatic stem and progenitor cells use distinct cell division strategies to generate differenti- ated cell types, and to renew the stem and pro- genitor pool. They can divide symmetrically to generate two equal cells that self-renew (symmetric self-renewing division) or differ- entiate (symmetric differentiating division). Alternatively, they can undergo asymmetric cell divisions (ACDs) to generate both self- renewing and differentiating progeny each time they divide (asymmetric self-sustaining divisions) (Fig. 1a). Symmetric self-renewing divisions amplify the stem cell compartment, whereas ACDs balance proliferation with dif- ferentiation and are used during functional development, such as neurogenesis, and in homeostatic tissue 1 . Through their ability to undergo both cell division modes, stem and progenitor cells are able to respond to micro- environment changes such as those induced by injury 2 . However, aberrant control of cell division mode carries a risk of enhanced tumorigenesis 1 . Our knowledge of ACD regulatory mecha- nisms largely originates from Drosophila melanogaster neuroblast studies 1 , where a stepwise process during mitosis leads to the establishment of an apicobasal polarity axis along which the mitotic spindle orients. This polarization allows cell fate determinants with opposing functions to localize to oppo- site poles and to segregate unequally during cytokinesis, which promotes self-renewal in one daughter cell and differentiation in the other. Although evidence exists for ACDs in mammalian stem and progenitor cells, and cancer stem cells 1 , how these integrate with regulatory mechanisms of adult mammalian tissue homeostasis and cancer is unclear. In this issue, Hwang et al. 3 demonstrate that sig- nals promoting epithelial-to-mesenchymal transition (EMT) in colorectal cancer cells also determine the cell division mode of colorectal cancer stem cells (CRCSCs). In the normal intestine, intestinal stem cells (ISCs) expressing Lgr5 (leucine-rich-repeat containing G-protein-coupled receptor 5), and located at the bottom of intestinal crypts, self-renew and generate transit-amplifying cells that expand, migrate upwards and dif- ferentiate at the crypt–villus junction 4 . The 212 NATURE CELL BIOLOGY VOLUME 16 | NUMBER 3 | MARCH 2014 © 2014 Macmillan Publishers Limited. All rights reserved

Transcript of A microRNA-operated switch of asymmetric-to-symmetric cancer stem cell divisions

Page 1: A microRNA-operated switch of asymmetric-to-symmetric cancer stem cell divisions

N E W S A N D V I E W S

a widely used diabetic mouse model in which β-cells are not able to compensate for the mas-sive need for insulin (BKS.db/db mice), islet SIK2 protein levels were actually reduced in spite of overt diabetes (and thus hyperglycae-mia), indicating that SIK2 stabilization (and potentially activity) must be controlled by alternative mechanisms — not only by glu-cose levels. Thus, finding upstream regulators of SIK2 activity will be important, as it might also reveal molecular mechanisms leading to β-cell failure in insulin-resistant or diabetic states. It remains to be tested if forced expres-sion or stabilization of SIK2 in β-cells could ameliorate the diabetic phenotype of the BKS.db/db mouse or other models of obesity that cannot compensate for their insulin resist-ance. The work by Screaton and colleagues further suggests that PJA2 plays a major role in β-cell physiology. PJA2 activity is known to

be modulated by intracellular cyclic adenosine monophosphate (cAMP) levels in vitro14, and cAMP levels in β-cells are actively regulated — for example, by the intestinal hormone GLP1 (glucagon-like peptide 1), whose long-lasting analogues are used as T2D therapeutics15. Whether SIK2 and PJA2 are co-expressed in human β-cells, and whether glucose-mediated stabilization of SIK2 occurs in this setting as well, remains to be tested experimentally. Furthermore, it will be interesting to examine if SIK2 also controls the secretion of other hor-mones regulating glucose homeostasis, includ-ing GLP1 secretion by intestinal L-cells.

This study of SIK2’s function in β-cells clearly advances our knowledge of β-cell physiology, and future investigations into the regulation of SIK2 function may be fruitful in the hunt for new therapeutic approaches for treatment of T2D.

COMPETING FINANCIAL INTERESTSThe authors declare no competing financial interests.

1. Konner, A. C. & Bruning, J. C. Cell Metab. 16, 144–152 (2012).

2. Ashcroft, F. M. & Rorsman, P. Cell 148, 1160–1171 (2012).

3. Sakamaki, J. et  al. Nat. Cell Biol. 16, 234–244 (2014).

4. Rathmann, W. & Giani, G. Diabetes Care 27, 2568–2569 (2004).

5. Lumeng, C. N. & Saltiel, A. R. J. Clin. Invest. 121, 2111–2117 (2011).

6. Bright, N. J., Thornton, C. & Carling, D. Acta Physiol. 196, 15–26 (2009).

7. Ruderman, N. B., Carling, D., Prentki, M. & Cacicedo, J. M. J. Clin. Invest. 123, 2764–2772 (2013).

8. Sasaki, T. et al. Neuron 69, 106–119 (2011).9. Bricambert, J. et al. J. Clin. Invest. 120, 4316–4331

(2010).10. Horike, N. et al. J. Biol. Chem. 278, 18440–18447

(2003).11. Kulkarni, R. N. et al. Cell 96, 329–339 (1999).12. Okada, T. et al. Proc. Natl Acad. Sci. USA 104, 8977–

8982 (2007).13. Wei, F. Y. et al. Nat. Med. 11, 1104–1108 (2005).14. Lignitto, L. et  al. Nat. Cell Biol. 13, 412–422

(2011).15. Seino, S. Diabetologia 55, 2096–2108 (2012).

Robin G. Lerner and Claudia Petritsch are in the Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Comprehensive Cancer Center, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California, USA. e-mail: [email protected]

A microRNA-operated switch of asymmetric-to-symmetric cancer stem cell divisionsRobin G. Lerner and Claudia Petritsch

Defective asymmetric cell divisions of stem and progenitor cells are associated with tumorigenesis by a largely unknown mechanism. A signalling axis involving Snail, microRNA-146a and Numb is now shown to regulate the switch between symmetric and asymmetric cell division in colorectal cancer stem cells.

Somatic stem and progenitor cells use distinct cell division strategies to generate differenti-ated cell types, and to renew the stem and pro-genitor pool. They can divide symmetrically to generate two equal cells that self-renew (symmetric self-renewing division) or differ-entiate (symmetric differentiating division). Alternatively, they can undergo asymmetric cell divisions (ACDs) to generate both self-renewing and differentiating progeny each time they divide (asymmetric self-sustaining divisions) (Fig. 1a). Symmetric self-renewing divisions amplify the stem cell compartment,

whereas ACDs balance proliferation with dif-ferentiation and are used during functional development, such as neurogenesis, and in homeostatic tissue1. Through their ability to undergo both cell division modes, stem and progenitor cells are able to respond to micro-environment changes such as those induced by injury2. However, aberrant control of cell division mode carries a risk of enhanced tumorigenesis1.

Our knowledge of ACD regulatory mecha-nisms largely originates from Drosophila melanogaster neuroblast studies1, where a stepwise process during mitosis leads to the establishment of an apicobasal polarity axis along which the mitotic spindle orients. This polarization allows cell fate determinants with opposing functions to localize to oppo-site poles and to segregate unequally during

cytokinesis, which promotes self-renewal in one daughter cell and differentiation in the other. Although evidence exists for ACDs in mammalian stem and progenitor cells, and cancer stem cells1, how these integrate with regulatory mechanisms of adult mammalian tissue homeostasis and cancer is unclear. In this issue, Hwang et al.3 demonstrate that sig-nals promoting epithelial-to-mesenchymal transition (EMT) in colorectal cancer cells also determine the cell division mode of colorectal cancer stem cells (CRCSCs).

In the normal intestine, intestinal stem cells (ISCs) expressing Lgr5 (leucine-rich-repeat containing G-protein-coupled receptor  5), and located at the bottom of intestinal crypts, self-renew and generate transit-amplifying cells that expand, migrate upwards and dif-ferentiate at the crypt–villus junction4. The

212 NATURE CELL BIOLOGY VOLUME 16 | NUMBER 3 | MARCH 2014

© 2014 Macmillan Publishers Limited. All rights reserved

Page 2: A microRNA-operated switch of asymmetric-to-symmetric cancer stem cell divisions

N E W S A N D V I E W S

cell division mode of ISCs has been intensely investigated, with one study reporting that about 40% of them divide in a perpendicular plane and generate progeny with distinct pro-liferation capacities, indicative of ACD (ref. 5). In contrast, clonal fate mapping experiments indicated that Lgr5+ cells divide symmetrically, and their progeny eventually acquire stem or transit-amplifying cell fate in a stochastic man-ner instructed by niche signals6. More recently, ISCs were found to partially switch from sym-metric, self-renewing division in infancy to asymmetric divisions in larger crypts7. These conflicting results could potentially be attrib-uted to different experimental settings. Taken together, the current research suggests that a sub-population of ISCs divides asymmetrically in Lgr5+ and Lgr5− progeny during a defined phase of crypt formation.

Disruption of ACD during the neoplastic transformation of mammalian stem and pro-genitor cells has been reported in premalignant oligodendrocyte progenitor cells, leukaemic progenitors and mammary stem cells, sug-gesting that ACDs act as a tumour suppressive mechanism by restricting proliferation and balancing it with differentiation1. However, whether ACD disruption is involved in colo-rectal cancer initiation is unclear. Colorectal-cancer-initiating mutations target and activate the Wnt pathway8, with Wnt-transformed ISCs identified as the adenoma cell of origin9. As asymmetrically dividing ISCs seem to be temporally restricted to early postnatal stages7, and tumours develop in adolescent and adult genetically engineered mice9, whether ISC transformation is accompanied by loss of ACD remains unknown.

Nevertheless, insights into the control of cell division mode have been provided by studies on CRCSCs, which are thought to be crucial for tumour growth and progression based on their increased tumour-initiating capacity in immunocompromised mice10. Notch signalling promotes growth of ISCs and colorectal cancer cells, and self-renewal of CRCSCs10. These stem cells can be identified by their sphere-forming capacity and slow proliferation rates in vitro, and were shown to distribute Numb asymmet-rically during division11. Numb is a conserved regulator of cell fate that inhibits Notch signal-ling, thereby promoting differentiation at the expense of self-renewal1. In CRCSCs, switch-ing from asymmetric to symmetric Numb distribution following depletion of ID1 and ID3 (inhibitor of DNA binding 1 and 3) was

shown to correlate with increased differentia-tion and decreased self-renewal11. Moreover, xenograft experiments with ID1/3-deficient CRCSCs associated symmetric differentiat-ing divisions with elevated chemosensitivity11. Further work demonstrated that CRCSCs isolated from late cancers predominantly undergo symmetric self-renewing divisions, and linked the control of division mode to microRNA (miR)-34a (ref. 12). This miRNA, which has tumour-suppressive activity in solid tumours13, was distributed asymmetrically to

the differentiating daughter cell in 10–20% of CRCSC divisions. miR-34ahigh cells downregu-lated Notch signalling and differentiated into a non-cancer stem cell, whereas the miR-34alow cells proliferated and self-renewed. These find-ings suggest that in early colorectal cancer, a cellular hierarchy headed by CRCSCs is estab-lished by consecutive asymmetric and sym-metric distribution of mi-R34a, and provide evidence that miRNA thresholds may exert tumour-suppressive functions by affecting binary cell fate decisions.

Wnt

WntNotch

Self-renewalDifferentiation

Asymmetric self-sustainingSymmetric self-renewing

Snail-induced EMTTumour growth

Asymmetric self-sustaining divisions

Intestinal stem cellEarly-stage CRC

Colorectal CSCsLate-stage CRC

Symmetric differentiatingba

d

c

Therapy resistance

Metastasis

Notch

E-cad

miR-146

Numb

β-cat

Symmetricself-renewing divisions

E-cad

β-catn

miR146a

Numb

Snail+TCF4

Wnt

Figure 1 Cell division modes used by cancer stem cells and their regulation in colorectal cancer. (a) Colorectal cancer stem cells (CRCSCs) undergo symmetric self-renewing divisions giving rise to two self-renewing daughter cells (red). The red curved arrows indicate self-renewal. (b) CRCSCs also undergo asymmetric self-sustaining divisions by localizing cell fate determinants with opposite functions (red and blue crescents) to opposite poles, thereby generating one self-renewing (red) and one differentiating (blue) daughter cell. (c) Symmetric differentiating divisions can also be induced in non-CSC colorectal cancer cells (blue); for example, by the presence of miR-34a (ref. 12). (d) Model of integrated regulation of cell division mode, tumour progression and metastasis. Left: miRNA-regulated Wnt and Notch signalling promotes symmetric self-renewing cell divisions of intestinal stem cells at the bottom of intestinal crypts. In non-neoplastic stem cells and CRCSCs from early tumour stages, Snail expression is low or absent, whereas Numb expression is high, leading to proteasomal degradation of β-catenin (β-cat) and attenuation of Wnt signalling. As a result, intestinal stem cells and early-stage CRCSCs divide asymmetrically. Right: Hwang et al.3 propose a positive feedback mechanism in late tumorigenesis, whereby the Snail-dependent action of nuclear β-catenin (β-catn) and TCF4 induces miR-146a expression and the subsequent downregulation of Numb. This relieves the Numb-mediated degradation of β-catenin and enhances Wnt signalling, thereby increasing symmetric divisions and maintaining the self-renewing CRCSC phenotype. The Snail–β-cat–miR-146a axis promotes tumour growth and metastasis, in part independently of the epithelial-to-mesenchymal transition (EMT).

NATURE CELL BIOLOGY VOLUME 16 | NUMBER 3 | MARCH 2014 213

© 2014 Macmillan Publishers Limited. All rights reserved

Page 3: A microRNA-operated switch of asymmetric-to-symmetric cancer stem cell divisions

N E W S A N D V I E W S

The work of Hwang et  al. now reveals another layer of miR- and Numb-dependent regulation of ACD in CRCSCs3. At late stages, tumour cells undergo EMT, a key devel-opmental program that, when activated in cancer, promotes invasion and metastasis14. EMT-inducing transcription factors, such as Snail, have been linked to increased stem-like phenotypes of mammary carcinoma cells14. Hwang et al. now provide mechanistic insights into how Snail increases stem cell properties. They observed that CRCSCs derived from established colorectal cancer cell lines predom-inantly underwent symmetric cell divisions, distributing Snail and the cancer stem cell marker CD44 equally between daughter cells. Following Snail depletion, CRCSCs switched to ACD, distributing CD44 asymmetrically and opposite to differentiation markers such as BMP4 (bone morphogenetic protein 4) and cytokeratin 20. Increased ACD rates correlated with restricted self-renewal capacity, indicating that Snail is critical for symmetric self-renew-ing divisions. miRNA expression analysis of CRCSCs revealed that miR-146a was upregu-lated by Snail, and further experiments dem-onstrated that the Snail-dependent induction of miR-146a was indirect, functioning through the activity of β-catenin and TCF4 (transcrip-tion factor 4), two key mediators of canonical Wnt signalling. Functional analyses showed that miR-146a acts as a major effector for the Snail-induced CRCSC phenotype. miR-146a depletion reduced self-renewal, clonogenicity and xenograft tumour growth, whereas ectopic expression of miR-146a enhanced these abili-ties without affecting cell proliferation or viabil-ity. Furthermore, although ectopic expression of miR-146a failed to affect migration and to induce complete EMT, its downregulation promoted asymmetric self-sustaining divisions and attenuated metastasis. By identifying that this Snail- and miR-146a-dependent signalling network influences cell division mode, Hwang et  al.3 show a divergence from the classical Snail-induced effects on EMT.

To address how miR-146a affects cell divi-sion mode, the authors combined computa-tional target predictions with gene expression data from miR-146a-overexpressing cells, and identified Numb as a major miR-146a target. They further delineated a positive feedback loop, whereby miR-146a maintains Wnt signal-ling by blocking Numb-dependent proteasomal degradation of β-catenin. Accordingly, enforced Numb downregulation enhanced Wnt signal-ling and increased symmetric self-renewing divisions, whereas restoring Numb levels in miR-146a-expressing cells had the opposite effect and reduced stem-cell-related properties in vitro and tumour growth in vivo. In line with the previously identified role of Notch in pro-moting symmetric self-renewing divisions12, the authors showed that pharmacological inhibition of Notch signalling in CRCSCs increased ACD rates. Unexpectedly, neither ectopic expression of Numb nor depletion of miR-146a repressed Notch signalling, indicating that Numb oper-ates in this CRCSC context predominantly by targeting β-catenin (Fig. 1b). This result might explain the puzzling observation that ectopic Numb expression, which presumably occurred in both daughter cells, led to unequal segrega-tion of Numb and restored ACD rather than induced symmetric differentiating divisions, as would be expected based on the function of Numb as a Notch inhibitor. The extent to which Numb promotes acquisition of differentiated fate in CRCSCs might be confounded by sev-eral factors, including isoform-specific effects, regulated phosphorylation status of Numb, or Numb- and Notch-independent regulation of ACD and cell fate — and will be important to address in future studies. Using an inhibitor that blocks Wnt activity downstream of EGF (epithelial growth factor), the authors further showed that pharmacological disruption of the Snail–miR-146a axis increased asymmetric self-sustaining divisions and attenuated tumour progression. They also observed that symmetri-cally expanding CRCSCs were more resistant to Cetuximab, a common inhibitor of the EGF

receptor. Finally, a SnailHighNumbLow expression profile was found to correlate with Cetuximab resistance and poor prognosis in colorectal can-cer patients.

The present study presents important mech-anistic insights into the regulation of symmet-ric and asymmetric cell divisions and the role of Snail in increasing CRCSC numbers to pro-mote tumour growth, and provides evidence that a cell division mode switch might contrib-ute to metastasis. However, several open ques-tions remain. For example, it is unclear how the Snail–miR-146a signalling axis and the associ-ated increased symmetric cell divisions might promote metastasis. Moreover, it would be important to dissect the precise role of Numb and the pathways it regulates, including Notch and Wnt, in the control of CRCSC fate. Based on the present3 and previous studies11,12, miR-NAs and ID1/3 emerge as important determi-nants of cell division mode, and it would be interesting to investigate whether these factors and the signalling networks they modulate also influence ACD in ISCs. In addition, as cancer stem cells are thought to mediate resistance to traditional therapies15, the present study raises the exciting possibility that pharmacological modulation of ACD rates could provide thera-peutic opportunities in colorectal cancer.

COMPETING FINANCIAL INTERESTSThe authors declare no competing financial interests.

1. Gómez-López, S., Lerner, R. G. & Petritsch, C. Cell Mol. Life Sci. 71, 575–597 (2013).

2. Troy, A. et al. Cell Stem Cell 11, 541–553 (2012).3. Hwang, W-L. et al. Nat. Cell Biol. 16, 268–280 (2014). 4. Clevers, H. Cell 154, 274–284 (2013).5. Quyn, A. J. et al. Cell Stem Cell 6, 175–181 (2010).6. Snippert, H. J. et al. Cell 143, 134–144 (2010).7. Itzkovitz, S., Blat, I. C., Jacks, T., Clevers, H. & van

Oudenaarden, A. Cell 148, 608–619 (2012).8. Kinzler, K.  W. & Vogelstein, B. Cell 87, 159–170

(1996).9. Barker, N. et al. Nature 457, 608–611 (2009).10. Miyamoto, S. & Rosenberg, D. W. Cancer Sci. 102,

1938–1942 (2011).11. O’Brien, C. A. et al. Cancer Cell 21, 777–792 (2012).12. Bu, P. et al. Cell Stem Cell 12, 602–615 (2013).13. Hermeking, H. Cell Death Differ. 17, 193–199 (2010).14. Tam, W. L. & Weinberg, R. A. Nat. Med. 19, 1438–49

(2013).15. Boman, B. M. & Huang, E. J. Clin. Oncol. 26, 2828–

2838 (2008).

214 NATURE CELL BIOLOGY VOLUME 16 | NUMBER 3 | MARCH 2014

© 2014 Macmillan Publishers Limited. All rights reserved