Vous êtes sur la page 1sur 3

Cell Stem Cell

Previews

1995). In fact, gene expression pro- 2010). In addition, the mutant cells differ- multiple diseases characterized by defec-
grams activated by cell-surface receptors entiate and thus exit from the stem cell tive stem cell differentiation, including
impinging on Rho proteins rely on the pool, resulting in defective wound closure cancer, or by accelerated stem cell deple-
nuclear integration of mechanisms sensing (Castilho et al., 2010). We can hypothesize tion, which leads to loss of tissue regener-
cytoskeletal changes with activation of that the requirement for the integration of ative capacity and premature aging.
MAPK cascades (Bar-Sagi and Hall, 2000; proliferative cues with mechanosensing
Posern and Treisman, 2006; Olson and pathways in order to communicate the REFERENCES
Nordheim, 2010). Thus, we can speculate condition of the niche may serve as a fail-
Bar-Sagi, D., and Hall, A. (2000). Cell 103, 227–238.
that if epithelial stem cells attach to the safe mechanism that triggers differenti-
ECM but are restricted from subsequent ating gene programs when stem cells Castilho, R.M., Squarize, C.H., Chodosh, L.A., Wil-
liams, B.O., and Gutkind, J.S. (2009). Cell Stem
spreading by constraints imposed by the sense aberrant biophysical conditions Cell 5, 279–289.
surface geometry, only partial deployment (Figure 1). The depletion of epithelial stem
of the integrin-initiated signaling network cells by promoting their differentiation in Castilho, R.M., Squarize, C.H., Leelahavanichkul,
K., Zheng, Y., Bugge, T., and Gutkind, J.S.
may result, and hence lead to the uncoordi- response to extrinsic or intrinsic cellular (2010). PLoS ONE 5, e10503.
nated activation of Rho GTPases, MAPKs, stress pathways may also act as a pro-
Connelly, J.T., Gautrot, J.E., Trappmann, B., Tan,
and cytoskeletal components. This combi- tective mechanism against oncogenic D.W., Donati, G., Huck, W.T., and Watt, F.M.
nation of signals may result in the initiation perturbation of this particularly vulnerable (2010). Nat. Cell Biol. 12, 711–718.
of cell differentiation programs, causing self-renewing cell population (Castilho Coso, O.A., Chiariello, M., Yu, J.C., Teramoto, H.,
cells to exit the cell cycle permanently and et al., 2009). Alternatively, this emerging Crespo, P., Xu, N., Miki, T., and Gutkind, J.S.
thus the stem cell pool. tendency of stem cells to undergo differen- (1995). Cell 81, 1137–1146.
Although the relevance of these mecha- tiation under stress conditions may also Ezhkova, E., Pasolli, H.A., Parker, J.S., Stokes, N.,
nisms to in vivo stem cell maintenance contribute to pathological events, such as Su, I.H., Hannon, G., Tarakhovsky, A., and Fuchs,
E. (2009). Cell 136, 1122–1135.
remains to be elucidated, it is known that accelerated tissue aging or reduced tissue
signaling events that mediate the interac- regeneration. On the other hand, failure to Geiger, B., Spatz, J.P., and Bershadsky, A.D.
tion between the basement membrane activate differentiation pathways may (2009). Nat. Rev. Mol. Cell Biol. 10, 21–33.

and the basal epidermal stem cells are contribute to psoriasis and other skin hy- Olson, E.N., and Nordheim, A. (2010). Nat. Rev.
required to maintain their stem-like state. perproliferative conditions. Hence, the Mol. Cell Biol. 11, 353–365.
For example, stem cells lacking the small molecular dissection of the underlying Posern, G., and Treisman, R. (2006). Trends Cell
GTPase Rac1 fail to undergo shape mechanisms that exist in the epidermal Biol. 16, 588–596.
changes and migrate on the provisional stem cell niche may provide new molecular Samuel, M.S., and Olson, M.F. (2010). Cell Stem
ECM at the wound edge (Castilho et al., targets for pharmacological intervention in Cell 7, this issue, 135–136.

Differential DNA Damage Response


in Stem and Progenitor Cells
Jun Seita,1,* Derrick J. Rossi,2,3,4,5 and Irving L. Weissman1
1StanfordInstitute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
2Harvard Stem Cell Institute, Boston, MA 02115, USA
3Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
4Immune Disease Institute, Boston, MA 02115, USA
5Program in Cellular and Molecular Medicine, Children’s Hospital Boston, Boston, MA 02115, USA

*Correspondence: jseita@stanford.edu
DOI 10.1016/j.stem.2010.07.006

The long lifespan of tissue-specific stem cells suggests that they may respond differently to DNA damage than
downstream cells. In this issue of Cell Stem Cell, two groups address this hypothesis by examining DNA
damage responses in hematopoietic stem and progenitor cells (Milyavsky et al., 2010; Mohrin et al., 2010).

Nearly 50 years after Till and McCulloch hematopoietic cell radiation sensitivity stem cell (HSC) radiosensitivity are
discovered clonal myeloerythroid cells (Till and McCulloch, 1961), the mecha- beginning to be unraveled. In contrast
in the pursuit of developing assays for nisms and the meaning of hematopoietic to somatic cells that are frequently

Cell Stem Cell 7, August 6, 2010 ª2010 Elsevier Inc. 145


Cell Stem Cell

Previews

postmitotic and/or short-lived, tissue- unclear whether the rescued cells convert when HSPCs were prestimulated into
specific stem cells must function DNA damage to mutations and/or translo- cycle. Taken together, these findings sug-
throughout life in order to mediate cations. These important issues have now gest that mouse HSC quiescence and reli-
ongoing tissue homeostasis and repair. been addressed in two papers published ance on NHEJ may be an important
The enormous functional demands and in this issue of Cell Stem Cell (Milyavsky mechanism contributing to mutagenesis
longevity of stem cells suggests that et al., 2010; Mohrin et al., 2010). The at the stem cell level that may serve as
stem cells, particularly those from highly studies used the hematopoietic stem the origin of certain blood cell cancers.
regenerative compartments such as the and progenitor compartments of mice In a complementary study published in
skin and blood, may be equipped with (Mohrin et al., 2010) and humans (Milyav- this issue, Milyavsky et al. addressed
effective DNA damage response (DDR) sky et al., 2010) to address the mecha- DDR by using hematopoietic stem and
mechanisms to ensure genomic integrity nisms through which stem and progenitor progenitor cells isolated from human
over a lifetime. With this said, it is also cells respond to DNA double-strand cord blood (CB). Strikingly, and in con-
increasingly clear that HSCs specifically breaks (DSBs) induced by ionizing trast to the murine study, irradiated
accumulate DNA damage during aging, radiation. human CB HSPCs exhibit a slower DNA
a mechanism believed to contribute to In the mice, Mohrin et al. addressed this damage repair kinetic and proved less
the elevated incidence of cancer in the issue by using hematopoietic stem and radio-resistant than downstream pro-
elderly (Rossi et al., 2007). progenitor cells (HSPCs) and myeloid pro- genitor cells. Knockdown of p53, a domi-
In the hematopoietic system, only genitors isolated from young mice. They nant form of p53, or ectopic expression
HSCs, memory T, and memory B lympho- found that at certain doses of ionizing irra- of BCL2 revealed that the p53-BCL2
cytes can self-renew; the hierarchical diation (2 Gy), HSPCs were more resistant pathway was involved in mediating the
differentiation scheme underlying HSC than downstream myeloid progenitors, apoptotic response of irradiated cord
lineage commitment makes DNA damage a result consistent with earlier studies blood progenitors. Knockdown and func-
accrual in HSCs a dangerous prospect. (Meijne et al., 1991). In the absence of tional evaluation of ASPP1, a molecular
In all stem cell-driven tissues, misrepaired ATM, all populations show equal radiation regulator of p53-dependent apoptosis,
lesions arising in stem cells can be propa- sensitivity, suggesting that ATM is suggested that stem cell radiosensitivity
gated both to daughter stem cells and involved in the differential DDR of HSPCs was at least in part mediated by ASPP1.
downstream progeny through the pro- versus progenitors. This pathway induces To further evaluate the functional conse-
cesses of self-renewal and differentiation. both cell-cycle arrest and expression of quence of DNA damage and response
In such a way, mutations arising in stem prosurvival genes in HSPCs, followed by on the function of CB HSPCs, the authors
cells can thus be potentiated throughout DNA repair. In contrast, damaged myeloid assessed their viability in the NOD/SCID
all cells of the tissue (Rossi et al., 2008). progenitors tended to be eliminated by repopulation assay. Irradiation (3 Gy)
Given that the cost of mutation accumula- apoptosis. The fact that quiescent HSPCs quantitatively abrogated the ability of
tion in stem cells is so high, it has been are almost uniformly in the G0 phase of cord blood cells to repopulate recipients,
postulated that stem cells may be the cell cycle suggested that they would which could be partially rescued by
uniquely privileged with properties that be obliged to use the error-prone nonho- knocking down p53 or overexpressing
either limit DNA damage accrual or effec- mologous end joining (NHEJ) pathway, BCL2. The authors then examined the
tively respond to it once damage is in contrast to the more high fidelity homol- affect of p53 and BCL2 on genomic integ-
incurred. Consistent with this model, ogous recombination (HR) pathway to rity during serial transplantation. Interest-
stem cells from diverse tissues possess repair DSBs. This hypothesis was tested ingly, DNA damage (as measured by
a variety of cytoprotective properties. by immunostaining and with a functional gH2AX foci) levels were similar in primitive
For example, stem cells from a variety of assay, which confirmed that HSPCs pref- hematopoietic progenitors expressing
tissues exhibit elevated ABC transporter erentially utilized NHEJ in contrast to either p53 or BCL2 in primary transplant
activity, resulting in an increased ability cycling progenitor cells. HSPCs driven recipients, yet in secondary recipients,
to pump genotoxic compounds out of into cycle prior to damage, however, p53 knockdown alone led to increased
the cell. Stem cells from several adult enabled access to HR to repair DSBs. indication of DNA damage accrual, which
tissues reside in a noncycling, quiescent Interestingly, HSPCs driven into cycle still correlated with a diminished capacity of
posture, minimizing the chance of rep- exhibited radioresistance over myeloid the p53-deficient cells to repopulate the
lication error (Yamazaki et al., 2006). progenitors. The reliance of quiescent secondary recipients. On the basis of
Moreover, quiescent stem cells are meta- HSPCs on the error-prone NHEJ pathway these findings, the authors propose that
bolically inactive and as such, generate raised the possibility that DNA damage the p53 apoptotic response is uncoupled
low levels of endogenous free radicals might be preferentially misrepaired in from DNA damage accumulation and
and reactive oxygen species (Tothova these cells. To test this point, the authors suggest an apoptosis-independent role
et al., 2007). irradiated cycling or quiescent HSPCs for p53 in human CB HSC self-renewal.
Less is known, however, about how and then examined genomic integrity in Collectively, the findings of Mohrin and
stem cells respond to DNA lesions once their progeny by spectral karyotyping. Milyavsky and their respective colleagues
they have happened. Overexpression of Strikingly, irradiated quiescent HSPCs have opened the door for a deeper
Bcl2 provides relative protection of gave rise to progeny that harbored chro- understanding of DDR in stem cells. Strik-
HSCs in terms of cell death responses mosomal abnormalities at a higher fre- ingly, a number of findings in these
(Domen and Weissman, 2003), but it is quency and in higher numbers than studies suggest that murine and human

146 Cell Stem Cell 7, August 6, 2010 ª2010 Elsevier Inc.


Cell Stem Cell

Previews

hematopoietic stem and progenitor to the stage of ontogeny from which Milyavsky, M., Gan, O.I., Trottier, M., Komosa, M.,
Tabach, O., Notta, F., Lechman, E., Hermans, K.
compartments may respond to DNA in the studied cells were derived, given G., Eppert, K., Konovalova, Z., et al. (2010). Cell
different ways. For example, the DNA that that both anatomical location and Stem Cell 7, this issue, 186–197.
damage repair kinetics of human CB ontogeny are known to significantly
HSPCs was delayed compared to down- impact HSC function (Rossi et al., 2008). Mohrin, M., Bourke, E., Alexander, D., Warr, M.R.,
Barry-Holson, K., Le Beau, M.M., Morrison, C.G.,
stream progenitors, in contrast to murine Because many hematopoietic disorders and Passegué, E. (2010). Cell Stem Cell 7, this
HSPCs, which exhibited a faster repair that stem from DNA damage accrual arise issue, 174–185.
kinetic. Similarly, whereas human HSPCs during aging, these results also stress the
were proapoptotic and more radiosensi- importance of examining DNA damage Rossi, D.J., Bryder, D., Seita, J., Nussenzweig, A.,
Hoeijmakers, J., and Weissman, I.L. (2007). Nature
tive than downstream progenitors, murine response and damage accrual during 447, 725–729.
HSPCs were more radioresistant ap- ontogeny and aging.
peared more antiapoptotic than progen- Rossi, D.J., Jamieson, C.H., and Weissman, I.L.
(2008). Cell 132, 681–696.
itor cells. Interestingly, a recent paper by ACKNOWLEDGMENTS
Blanpain and colleagues examining the Sotiropoulou, P.A., Candi, A., Mascre, G.,
DNA damage response in murine hair I.W. owns significant stock in Amgen, Inc., from De Clercq, S., Youssef, K.K., Lapouge, G., Dahl,
past service on its SAB. He is a director and E., Semeraro, C., Denecker, G., Marine, J.C.,
follicle stem cells reported apoptotic and consultant and holds stock in Stem Cells, Inc., et al. (2010). Nat. Cell Biol. 12, 572–582.
repair kinetic properties more similar to and he is a former director and consultant and
murine HSPCs, suggesting that species- stockholder with Cellerant, Inc. He has consulted Till, J.E., and McCulloch, C.E. (1961). Radiat. Res.
specific differences may underlie these for Fate Therapeutics. 14, 213–222.
different findings (Sotiropoulou et al.,
REFERENCES Tothova, Z., Kollipara, R., Huntly, B.J., Lee, B.H.,
2010). Castrillon, D.H., Cullen, D.E., McDowell, E.P.,
Perhaps the most interesting possibility Lazo-Kallanian, S., Williams, I.R., Sears, C., et al.
Domen, J., and Weissman, I.L. (2003). Exp. Hema-
is that DDR differences seen in CB HSPCs (2007). Cell 128, 325–339.
tol. 31, 631–639.
from human versus bone marrow or
Meijne, E.I., van der Winden-van Groenewegen, R. Yamazaki, S., Iwama, A., Takayanagi, S., Morita,
mobilized HSPCs from young mice relate J., Ploemacher, R.E., Vos, O., David, J.A., and Y., Eto, K., Ema, H., and Nakauchi, H. (2006).
to the different anatomical locations, or Huiskamp, R. (1991). Exp. Hematol. 19, 617–623. EMBO J. 25, 3515–3523.

Cell Stem Cell 7, August 6, 2010 ª2010 Elsevier Inc. 147

Vous aimerez peut-être aussi