A Switch in Time Through Genes Aligned Unraveling The Geno - 2020 - Cell Stem

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 3

ll

Previews
and is an inventor on multiple patients related to Nicotinamide Metabolism Mediates Resistance to Pollyea, D.A., Stevens, B.M., Jones, C.L., Winters,
CD47 cancer immunotherapy licensed to Gilead. Venetoclax in Relapsed Acute Myeloid Leukemia A., Pei, S., Minhajuddin, M., D’Alessandro, A.,
Stem Cells. Cell Stem Cell 27, this issue, 748–764. Culp-Hill, R., Riemondy, K.A., Gillen, A.E., et al.
(2018). Venetoclax with azacitidine disrupts energy
REFERENCES Lagadinou, E.D., Sach, A., Callahan, K., Rossi, metabolism and targets leukemia stem cells in pa-
R.M., Neering, S.J., Minhajuddin, M., Ashton, tients with acute myeloid leukemia. Nat. Med. 24,
DiNardo, C.D., Pratz, K.W., Letai, A., Jonas, B.A., J.M., Pei, S., Grose, V., O’Dwyer, K.M., et al. 1859–1866.
Wei, A.H., Thirman, M., Arellano, M., Frattini, (2013). BCL-2 inhibition targets oxidative phos-
M.G., Kantarjian, H., Popovic, R., et al. (2018). phorylation and selectively eradicates quiescent Pollyea, D.A., Amaya, M., Strati, P., and
Safety and preliminary efficacy of venetoclax with human leukemia stem cells. Cell Stem Cell 12, Konopleva, M.Y. (2019). Venetoclax for AML:
decitabine or azacitidine in elderly patients with 329–341. changing the treatment paradigm. Blood Adv. 3,
previously untreated acute myeloid leukaemia: a 4326–4335.
non-randomised, open-label, phase 1b study. Pan, R., Hogdal, L.J., Benito, J.M., Bucci, D., Han,
Lancet Oncol. 19, 216–228. L., Borthakur, G., Cortes, J., DeAngelo, D.J.,
Debose, L., Mu, H., et al. (2014). Selective BCL-2 Thomas, D., and Majeti, R. (2017). Biology and
Jones, C.L., Stevens, B.M., D’Alessandro, A., inhibition by ABT-199 causes on-target cell death relevance of human acute myeloid leukemia stem
Reisz, J.A., Culp-Hill, R., Nemkov, T., Pei, S., in acute myeloid leukemia. Cancer Discov. 4, cells. Blood 129, 1577–1585.
Khan, N., Adane, B., Ye, H., et al. (2018). 362–375.
Inhibition of Amino Acid Metabolism Selectively Zhang, H., Nakauchi, Y., Köhnke, T., Stafford, M.,
Targets Human Leukemia Stem Cells. Cancer Pei, S., Pollyea, D.A., Gustafson, A., Stevens, B.M., Bottomly, D., Thomas, R., Wilmot, B.,
Cell 34, 724–740.e4. Minhajuddin, M., Fu, R., Riemondy, K.A., Gillen, McWeeney, S.K., Majeti, R., and Tyner, J.W.
A.E., Sheridan, R.M., Kim, J., et al. (2020). (2020). Integrated analysis of patient samples iden-
Jones, C.L., Stevens, B.M., Pollyea, D.A., Culp- Monocytic Subclones Confer Resistance to tifies biomarkers for venetoclax efficacy and com-
Hill, R., Reisz, J.A., Nemkov, T., Gehrke, S., Venetoclax-Based Therapy in Patients with Acute bination strategies in acute myeloid leukemia. Nat.
Gamboni, F., Krug, A., Winters, A., et al. (2020). Myeloid Leukemia. Cancer Discov. 10, 536–551. Can. 1, 826–839.

A ‘‘Switch’’ in Time through Genes Aligned:


Unraveling the Genomic Landscape
of HSC Development
April C. Apostol,1 Diego A. López,2,* and Anna E. Beaudin3,*
1Quantitative and Systems Biology Graduate Program, University of California-Merced, Merced, CA, USA
2Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
3Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake

City, UT, USA


*Correspondence: diego.lopez@path.utah.edu (D.A.L.), anna.beaudin@hsc.utah.edu (A.E.B.)
https://doi.org/10.1016/j.stem.2020.10.011

Seeking to define the ‘‘switch’’ from fetal to adult hematopoiesis, Li et al. (2020) performed extensive genomic
and epigenomic profiling of hematopoietic stem and progenitor cells across ontogeny (as explored in this
issue of Cell Stem Cell). Gradual and stochastic changes in genomic and epigenomic regulation suggest
the absence of any specific regulator.

Hematopoiesis is driven by HSCs (he- sets of innate-like lymphocytes, such as the neonatal period (Bowie et al., 2007),
matopoietic stem cells) and HPCs (he- peritoneal B-1a cells (Ikuta et al., 1990; neither the origins nor the temporal
matopoietic progenitor cells). The first Kikuchi and Kondo, 2006). These differ- mechanisms that govern this switch
definitive HSCs arise in the early embryo, ences can also be observed in have been ascertained. In this issue of
and are capable, like adult HSCs, of re- comparing the distinct transcriptional Cell Stem Cell, Li et al. profile the tran-
constituting adult hematopoiesis (Mu €ller profiles and dependencies of fetal and scriptional and epigenomic landscape
et al., 1994). Fetal HSCs, however, are adult HSCs (Copley and Eaves, 2013) of HSCs and HPCs across ontogeny to
still phenotypically and functionally but the mechanisms that drive the transi- gain insight into the genetic drivers of
disparate from adult bone marrow (BM) tion from fetal to adult phenotypes are this transition (Li et al., 2020).
HSCs (Copley and Eaves, 2013). They under investigation. While previous To approach the question of how the
are less quiescent and more proliferative, studies have described the ‘‘switch’’ in transition from fetal to adult identity oc-
yet display higher self-renewal potential HSC phenotype from fetal to adulthood curs, the authors performed single-cell
(Bowie et al., 2007). They also generate as the downregulation of fetal genes sequencing (scRNAseq) of sorted HSCs/
unique lineages including distinct sub- and upregulation of adult genes during HPCs across ontogeny, at E16.5, P7,

Cell Stem Cell 27, November 5, 2020 ª 2020 Published by Elsevier Inc. 695
ll
Previews

P14, and 8 weeks of age. Perhaps surpris- while repressive histones (H3K27me3) HPCs (and to a lesser extent, HSCs) was
ingly, neonatal HSCs/HPCs clustered were present on adult promoters and en- delayed in both scRNA and ATAC-seq ex-
independently from either fetal or adult hancers within fetal HSC/HPCs, there periments compared to control, due to
HSCs/HPCs, suggesting that the transi- were no differences in H3K27me3 levels Ifnar dependence of several adult identity
tion between the two identities is not on fetal promoters and enhancers within genes. Despite this delay, however, HSCs
bimodal, but rather gradual. Using a adult HSCs/HPCs, indicating that fetal otherwise retained normal function. These
quadratic programming approach, the au- identity is overwritten by adult program- observations suggest that IFN signaling
thors determined identity scores for fetal, ming. These epigenetic changes mirror coincides with this transition, but whether
neonatal, and adult HSCs/HPCs by inte- observations in transcriptomic data, it directly regulates the transition from
grating previously published datasets en- further emphasizing the successive and fetal to adult hematopoiesis remains to
riched for both fetal and adult identity uncoordinated changes between fetal be determined.
genes. In agreement with the concept of and adult HSC/HPC identity. Although the authors showcased exten-
a gradual transition, there was a progres- To probe the potential role of environ- sive profiling of HSCs across ontogeny us-
sive shift in expression of adult identity mental cues in the regulation of fetal-to- ing rich datasets that contribute greatly to
genes over time in fetal to adult clusters. adult transition, the investigators posited our current understanding of hematopoi-
Furthermore, these results were recapitu- that gradual changes to transcription etic development, the mechanisms that
lated using an unsupervised approach and chromatin accessibility during the drive these changes are still not entirely
that calculated identity scores of all ex- neonatal period may be initiated by the clarified. It is possible that gradual shifts
pressed genes, demonstrating that glob- migration of fetal HSCs from fetal liver in gene expression and chromatin acces-
ally, fetal and neonatal HSC and HPC (FL) to BM (Mu €ller et al., 1994). To test sibility reflect a fundamental shift in the
populations were becoming more adult- this, they compared single-cell transcripts makeup of HSCs and HPCs at different
like with age. of HSCs in P0 FL and BM to transcripts of time points, i.e., the changes observed
The authors proposed three possible HSCs in E16.5 FL and P7 BM. P0 HSCs are not within the same population of cells
models to explain the gradual changes in clustered together regardless of location, but are within temporally distinct cell pop-
HSC and HPC identity: (1) individual cells rather than with E16.5 or P7 HSCs, indi- ulations that arise in ‘‘waves’’ across
may coordinate global changes in gene cating that BM migration alone does not development. This view is supported by
expression through strictly regulated net- regulate establishment of adult identity. the observation that the Ifnar / pheno-
works; (2) environmental cues may direct Coinciding with the perinatal FL to BM type delayed the emergence of specific
coordinated changes in gene expression; transition, the robust and sudden expres- Flk2+ progenitors. Because FL hemato-
and (3) HSCs/HPCs may upregulate adult, sion of type I interferon (IFN) target genes, poiesis is marked by rapid expansion
and downregulate fetal, identity genes in such as Irf7, yielded another possible and differentiation, and HSCs with devel-
an uncoordinated manner. Analysis of driver of the switch between fetal to adult opmentally restricted activity exist along-
fetal and adult identity gene expression identity. Bulk-sequencing pinpointed the side canonical HSCs (Beaudin et al.,
within HSCs/HPCs across all three stages induction of IFN activity beginning at 2016), alterations to sterile signaling may
revealed that, although most cells identi- E16.5, with the highest expression occur- impinge upon these normal dynamics.
fied by higher adult identity scores aligned ring between E18.5 and P0, just prior to Furthermore, the phenotypic markers
with adult identity gene expression, they and after birth. IFNa expression in E18.5 used to characterize fetal HSC and HPC
also expressed fetal identity genes FL was 3-fold higher relative to earlier populations (Pietras et al., 2015) are not
(Igf2bp2, Hmga2, and Arid3a). In a similar time points. The spike in IFN could not as well-defined as their adult counterparts
fashion, cells with greater fetal identity be explained by exogenous maternal cy- and may not capture the true depth of HSC
scores also displayed low levels of adult tokines or a drop in progesterone in late heterogeneity. It may therefore be difficult
identity gene expression (Cpne2, H2-Q7, gestation. Normal fetal gut flora, which to parse out mechanistic differences from
and Sdsl). This observation is consistent expands between E16 and E18, was phenotype-sorted populations alone; a
with a model in which fetal identity genes also not the source, as evidenced by broader look at the entire hematopoietic
are gradually switched off as adult identity normal levels of IFN expression in HSCs/ stem and progenitor compartment may
genes are upregulated in an independent, HPCs at E18 in germ-free mice. RT-PCR be insightful. Together, these data provide
stochastic manner. of IFNa expression within fetal organs re- elegant insight into how HSCs transition
To gain more clarity on the processes vealed high expression in E18 skin into adulthood and also reveal how much
that govern gradual changes in HSC iden- compared to other fetal organs such as still remains to be understood about the
tity, the authors probed epigenetic regula- placenta and thymus. This suggests that complex nature and dynamics of hemato-
tion in the transition to adult identity. the skin may be a primary source of poietic development.
Similar to transcriptional analysis, ATAC IFNa, although further investigation is
sequencing revealed a steady shift needed. Analysis of Ifnar / mice re-
REFERENCES
from fetal to adult-like chromatin accessi- vealed that this perinatal spike in IFN pro-
bility across ontogeny. Although adult moted HSC expansion, while lack of IFNa Beaudin, A.E., Boyer, S.W., Perez-Cunningham,
enhancer regions became more open as receptor significantly depleted HPC pop- J., Hernandez, G.E., Derderian, S.C., Jujjavarapu,
the HSCs/HPCs matured, fetal identity ulations, particularly the lymphoid biased C., Aaserude, E., MacKenzie, T., and Forsberg,
E.C. (2016). A Transient Developmental
genes remained accessible. Through Flk2+ HPC-1 (MPP4). Not surprisingly, Hematopoietic Stem Cell Gives Rise to Innate-
ChIPmentation, they determined that the onset of adult identity in Ifnar / like B and T Cells. Cell Stem Cell 19, 768–783.

696 Cell Stem Cell 27, November 5, 2020


ll
Previews
Bowie, M.B., Kent, D.G., Dykstra, B., McKnight, potential occurs at the level of hematopoietic Reprogramming that Begins before Birth. Cell
K.D., McCaffrey, L., Hoodless, P.A., and Eaves, stem cells. Cell 62, 863–874. Stem Cell 27, this issue, 732–747.
C.J. (2007). Identification of a new intrinsically
timed developmental checkpoint that reprograms Mu€ller, A.M., Medvinsky, A., Strouboulis, J.,
Kikuchi, K., and Kondo, M. (2006). Developmental
key hematopoietic stem cell properties. Proc. Grosveld, F., and Dzierzak, E. (1994).
switch of mouse hematopoietic stem cells from
Natl. Acad. Sci. USA 104, 5878–5882. Development of hematopoietic stem cell activity
fetal to adult type occurs in bone marrow after
in the mouse embryo. Immunity 1, 291–301.
birth. Proc. Natl. Acad. Sci. USA 103,
Copley, M.R., and Eaves, C.J. (2013). 17852–17857. Pietras, E.M., Reynaud, D., Kang, Y.A., Carlin, D.,
Developmental changes in hematopoietic stem Calero-Nieto, F.J., Leavitt, A.D., Stuart, J.M.,
cell properties. Exp. Mol. Med. 45, e55. Li, Y., Kong, W., Yang, W., Patel, R.M., Casey, Göttgens, B., and Passegué, E. (2015).
E.B., Okeyo-Owuor, T., White, J.M., Porter, S.N., Functionally Distinct Subsets of Lineage-Biased
Ikuta, K., Kina, T., MacNeil, I., Uchida, N., Peault, Morris, S.A., and Magee, J.A. (2020). Single-Cell Multipotent Progenitors Control Blood Production
B., Chien, Y.H., and Weissman, I.L. (1990). A devel- Analysis of Neonatal HSC Ontogeny Reveals in Normal and Regenerative Conditions. Cell
opmental switch in thymic lymphocyte maturation Gradual and Uncoordinated Transcriptional Stem Cell 17, 35–46.

Dynamic Hydrogels
for Investigating Vascularization
Gordana Vunjak-Novakovic1,*
1Columbia University, New York, NY 10032, USA

*Correspondence: gv2131@columbia.edu
https://doi.org/10.1016/j.stem.2020.10.009

Extracellular matrix is known to regulate vascularization by a sequence of multiple factors including mechan-
ical forces. In this issue of Cell Stem Cell, Wei et al. (2020) investigate the roles of matrix biomechanics on
early stages of vasculogenesis by using hydrogels with tunable stiffness and stress relaxation.

Cell fate and function, in vivo and in vitro, tissue culture. While vascular phenotypes field has also started to incorporate
are regulated by the entire context of the change with the developmental stage, another facet of control where materials
cellular environment, through cascades parent tissue type, and the state of health that change their properties in response
of interacting molecular, structural, and or disease, achieving angiogenesis and to cell-generated signals are being
physical signals that change in space vasculogenesis is critically important explored. The present study is an impor-
and time. The cells in turn regulate their both for the survival of implanted tissues tant step toward establishing and utilizing
environment as they secrete cytokines, and for designing tissue models for bio- dynamic cellular environments. The au-
build and degrade the extracellular ma- logical research. In this study, the focus thors explored if the activation of specific
trix, influence the surrounding cells, and is on the effects of the mechanical envi- mechanosensing mechanisms in cultured
generate physical forces. These two-way ronment, established by a hydrogel used vascular cells, in conjunction with matrix
interactions between the cells and their to encapsulate cells, on the initiation, pro- remodeling, can be utilized to induce the
environment are instrumental for native gression, and outcomes of early stages of assembly of vascular networks. To this
tissue development, phenotypic changes vasculogenesis. end, they developed a viscoelastic hydro-
associated with diseases, and the assem- It is well known that the extracellular gel with dynamic crosslinks that pro-
bly and function of engineered tissues. In matrix provides not only a three-dimen- moted cellular contractility and enabled
recent years, in vitro models of increas- sional setting for the cultured cells but formation of large focal adhesions and
ingly high biological fidelity have been also serves as a major source of regulato- robust vascular networks. This sequence
developed to enable controllable studies ry signals that determine vascular of events could not be initiated using
under conditions more closely emulating development and function, including otherwise identical hydrogels with static
the in vivo milieu (Paek et al., 2019; Ro- physical forces (Discher et al., 2009). covalent crosslinks.
naldson-Bouchard and Vunjak-Nova- With this understanding, the field has To decouple the effects of stiffness and
kovic, 2018; Skylar-Scott et al., 2019). gradually moved from generic, inert, matrix dynamics, the authors conducted
In this issue of Cell Stem Cell, the Wei and nondegradable biomaterials toward experiments with dynamic and static
et al. (2020) article reports a study of designing those with tunable properties matrices that had the same initial stiffness
vascularization, which remains one of that can be tailored to control cell differen- at two different levels: low (soft matrix)
the universal challenges in cell and tiation and assembly. In recent years, the and high (stiff matrix). Cultivation of

Cell Stem Cell 27, November 5, 2020 ª 2020 Elsevier Inc. 697

You might also like