Sunteți pe pagina 1din 9

BBA - Molecular Cell Research 1866 (2019) 409–417

Contents lists available at ScienceDirect

BBA - Molecular Cell Research


journal homepage: www.elsevier.com/locate/bbamcr

Review

Cell size homeostasis: Metabolic control of growth and cell division T


Mikael Björklund
Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, Zhejiang University School of Medicine, International Campus, 718 East Haizhou Rd., Haining, Zhejiang
314400, PR China

A R T I C LE I N FO A B S T R A C T

Keywords: Joint regulation of growth rate and cell division rate determines cell size. Here we discuss how animal cells
Cell size control achieve cell size homeostasis potentially involving multiple signaling pathways converging at metabolic reg-
Metabolism ulation of growth rate and cell cycle progression. While several models have been developed to explain cell size
Growth rate control, comparison of the two predominant models shows that size homeostasis is dependent on the ability to
Cell size sensing
adjust cellular growth rate based on cell size. Consequently, maintenance of size homeostasis requires that larger
cells can grow slower than small cells in relative terms. We review recent experimental evidence showing that
such size adjustment occurs primarily at or immediately before the G1/S transition of the cell cycle. We further
propose that bidirectional feedback between growth rate and size results in cell size sensing and discuss potential
mechanisms how this may be accomplished.

1. Introduction results in a quite substantial 33% increase in cell volume. A further


complication is that the term ‘growth rate’ is often used to describe the
How cell size is determined and regulated is an important yet poorly increase in cell number per unit time. Instead, a more relevant measure
understood question in biology. While it appears that cells attempt to for cell size studies, and also used in this review, is to define growth rate
maintain their characteristic size, the mechanisms have remained elu- as the cell volume or mass increase per time (e.g., femtoliter/hour or
sive [1,2]. This review will concentrate on how proliferating animal picogram/hour).
cells control their size as research has primarily focused on how to Current understanding is that cell size is determined by a number of
achieve size homeostasis through regulation of cell growth and cell internal and external factors, e.g., genome size, nutrients and growth
division. This joint regulation of cell size is perhaps best illustrated factors [1,2], which affect cellular signaling pathways and metabolic
through selective inhibition of cell cycle progression. Under such con- activity. Changes in cell size are typically observed when the balance
ditions cells continue to grow largely uninterrupted [3,4] and may between the growth rate and cell division rate is altered. This balance is
reach sizes that would not be attainable under physiological conditions. a key determinant of cell size across different organisms from uni-
Therefore, both cell growth and normal cell cycle progression are re- cellular bacteria to multicellular animals. However, an important con-
quired for the maintenance of cell size homeostasis in proliferating ceptual problem arises from this observation. Namely, if cell size is
cells. The principles of how non-dividing cell types maintain their size determined by growth rate and cell cycle progression, it is easy to view
are less well established [1], which is unfortunate as non-dividing cells cell size a spandrel, an evolutionary byproduct [5]. Nevertheless, it is
constitute the great majority of cell types in the animal body. obvious from our macroscopic world that size has a huge impact on
Before going into any details of cell size regulation, it is important to organisms [6], e.g., just compare the structural and functional differ-
realize that many different parameters are used as a measure for cell ences between mice and elephants. Does size also matter for cells?
size, including cell length, area, volume, total cell mass, dry mass and Experiments suggest that cell size is important for cellular fitness and
total protein content. Typically, which one is used depends on selected function [7,8] and altered cell size appears to have diverse effects at the
techniques, available instrumentation and even individual preferences. whole animal level from brain function to aging [9,10]. Loss of cell size
In particular, cell length, area and volume are often used almost in- homeostasis is also commonly seen in chronic diseases such as cancers
terchangeably without consideration to the geometric implications. For [1,2].
example, while many investigators might overlook the significance of a Many general processes are size dependent, including transcription
10% increase in the diameter of a spherical-shaped cell, this actually [11,12], translation [13] and metabolism [12]. Additionally, specific

Abbreviations: SV, surface-to-volume; mTOR, mechanistic target of rapamycin; MAPK, mitogen-activated protein kinase
E-mail address: mikael.bjorklund.lab@gmail.com.

https://doi.org/10.1016/j.bbamcr.2018.10.002
Received 9 August 2018; Received in revised form 25 September 2018; Accepted 3 October 2018
Available online 11 October 2018
0167-4889/ © 2018 The Author. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
M. Björklund BBA - Molecular Cell Research 1866 (2019) 409–417

processes including actin filament and focal adhesion organization, increasing amount of single cell data clearly points to the conclusion
nuclear shape and cell contractility have also been shown to be influ- that the linear vs. exponential growth debate should finally put to an
enced by cell size [14]. All this data implies that cells adjust their end and replaced by more realistic, albeit more complex, models on
function to match their size and thus are somehow aware of their how cells grow.
physical dimensions. Such ‘size sensing’ could be an important me-
chanism for the maintenance of appropriate growth rate and/or cell 3. The main models for the maintenance of size homeostasis are
cycle progression [15,16]. Alternatively, size sensing could be directly not mutually exclusive
required for the control of cell size to maintain optimal cellular fitness
and function as suggested by the loss of cell size uniformity in many 3.1. ‘Sizer’, ‘timer’ and ‘adder’ models
cancers [1,2]. These explanations may not be mutually exclusive and
both interpretations imply that cell size matters. While the linear vs. exponential models have been used to argue for
and against the need for cell size control, several models have been
2. How do animal cells grow? The linear vs. exponential growth developed to explain how cell size homeostasis is maintained. These
debate include the ‘sizer’, ‘timer’ and ‘adder’ models. The ‘sizer’ assumes that a
cell needs to reach a critical size before it can divide, whereas the
In order to understand how cells maintain their size, it is important ‘timer’ suggests that cells grow for a certain time prior to dividing. The
to study how cells grow. In recent years, research efforts have con- ‘adder’ model proposes that cells add a constant volume during each
centrated on understanding cell size control mechanisms at a phe- cell cycle regardless of their size [27]. The ‘timer’ model maintains size
nomenological level and not so much at the level of molecular me- homeostasis if cells grow linearly for a constant amount of time.
chanisms [17,18]. This largely stems from the decades old debate However, this model is conceptually the most challenging one as it
whether cell growth is exponential or linear during the cell cycle assumes that cells have a mechanism to measure time. For this reason,
[4,19]. While this might sound trivial, there is a solid theoretical ar- the ‘timer’ model has lost much of its shine and will not be discussed
gument why this is important and experimental reasons why this debate further. However, it has been reported that growth during S and G2
continues to date. Theoretically, if cell growth is linear (larger and phases of the cell cycle can resemble timer behavior in some cell lines
smaller cells grow at a similar rate), population size homeostasis can be [23].
maintained without paying specific attention to cell size. However, if The ‘sizer’ model proposes that cells need to reach a minimal size in
growth is exponential (larger cells grow faster than smaller cells) then order to progress through G1/S transition or mitosis (Fig. 1A). This has
cell size needs to be regulated in order to maintain size homeostasis. lead a number of groups to propose that cells from bacteria to animal
This is because exponential growth causes size variance in the cell cells can sense their size [15,16,28,29]. Recent work by Ginzberg et al.
population to increase resulting in a loss of a stable cell size distribu- showed that HeLa and retinal pigment epithelial (RPE) cells adjust their
tion. Therefore, it is thought that exponential growth requires specific growth rates twice in each cell cycle [16]. Specifically, growth rate was
cell size checkpoints, which ensure that only cells that have reached a shown to be selectively increased in small cells and reduced in large
certain minimal size may progress uninterrupted through the cell cycle. cells in order to maintain cell size homogeneity. It is important to note
One of the practical reasons why the exponential or linear growth that many biological phenomena, including cell growth and metabo-
debate has continued so long is that it is still technically challenging to lism are multiplicative in nature, and consequently proportional rather
reliably measure the difference between linear and exponential growth than absolute changes are more noteworthy [30]. Therefore, growth
as these models differ maximally by less than 6% during a single cell rate changes discussed in this review should be understood in relative
cycle [20,21]. While such a high resolution is now achievable in single (the change per cell volume/mass) rather than absolute terms.
cell mass measurements [21,22], other factors affecting experimental The strongest adjustment of cell size variability [16] roughly cor-
variability, such as heterogeneity in growth rate of individual cells, are responds to the G1/S transition, consistent with classical models of
often larger than this. Furthermore, the kinetics of cell growth appears growth rate regulation at G1/S transition or immediately before at so
more complex than anticipated by these reductionistic models. Ex- called restriction point [31]. The authors pointed out that cell size
periments with animal cells have shown than growth rate is not con- variance can only decrease if small cells grow faster than large cells
stant during the cell cycle [16,20,23], and is not purely linear or ex- [16], a condition which counteracts the size homeostasis destabilizing
ponential [22]. Such cell-cycle dependent changes in growth rate are effect of exponential growth. Analysis of cell size variability as a
also evident in both budding yeast and fission yeast [24,25]. function of cellular DNA content as measured by flow cytometry sup-
Growth rate in animal cells can also be adjusted conditionally, de- ports the data from single cell imaging (Fig. 2). Finally, Ginzberg et al.
pending for example on cell size at birth (the cell size immediately after concluded that the cell's target size may be achieved through regulation
cell division) [26]. By studying single cell growth trajectories, Cadart of cyclin dependent kinase CDK4, a key regulator of cell cycle pro-
et al. recently found that while overall growth is exponential in G1 gression in G1 phase, highlighting the tight connection between cell
phase of the cell cycle, further growth is adjusted in S-G2 phases in the size and cell cycle progression. The requirement for CDK4 in this pro-
cell cycle so that there is a negative correlation between added volume cess is consistent with the recent suggestion that CDK4 regulates G1
in G1 and S-G2 [23]. Interestingly, this finding may relate to the ex- length [32]. Interestingly, while CDK4 was required for setting the
periments conducted by Conlon et al. [3,4], which revealed linear target size, cell size variance was found to be regulated by p38 MAPK
growth in Schwann cells, a neuronal cell type that produces the myelin kinase activity [33]. While this suggests that these processes are in-
sheath around axons. However, Conlon and Raff used cells arrested in S dependently regulated, p38 MAPK has also been linked to direct reg-
phase using a DNA replication inhibitor aphidicolin. Consequently, if ulation of CDK4 activity [34]. These encouraging results show how
growth rate changes observed by Cadart et al. are applicable to growth rate may be adjusted in conjunction with G1 length regulation.
Schwann cells, the use of aphidicolin would have resulted in missing Identification of further molecular details should help to reveal me-
the exponential growth phase in G1. This realization may therefore chanisms how cells can measure their size. For example, p38 MAPK
undermine some of the strongest evidence supporting linear growth. pathway is regulated by various signals including growth factors as well
Interestingly, these findings also suggest that the increased amount of as oxidative and osmotic stress. Which of these generates the signal
DNA is not accelerating growth, indicating that DNA template amount which p38 MAPK uses for regulation of cell size variance?
is not limiting growth during normal cell cycle progression. The high Accumulation of extensive single-cell time-lapse imaging data from
biosynthetic cost for genome duplication could at least in part explain bacteria to mammalian cells has also resulted in resurrection and re-
this growth rate adjustment in S-G2. Altogether, analysis of the finement of the ‘adder’ or ‘incremental’ model as it was initially called

410
M. Björklund BBA - Molecular Cell Research 1866 (2019) 409–417

A C
'Sizer' Volume Growth rate
increase

S= x
Cell volume

Size Cell cycle length

ADDER MODEL SIZE SENSING MODEL

Size added is constant Target size is constant


Large cells grow
slower and/or their cell
cycle is faster Size control
achieved by joint regulation of
growth and cell cycle
Division and/or
Birth
Time G1-S transition

B D
'Adder' Volume
increase

Metabolic rate Mass3/4


Cell volume

KLEIBER'S LAW

Metabolic activity is proportional to mass

Expected to reduce relative growth rate in larger


cell consistent with both adder and size sensing model
Relative growth rate of
large cells is slower
and/or cell cycle is faster

Birth Division
Time
Fig. 1. The ‘sizer’ and ‘adder’ models. (A) With ‘sizer’ cells need to reach certain size before they can start DNA synthesis (move through G1/S transition) or divide
and therefore cells that are initially of different sizes show different volume increase. (B) In the ‘adder’ model, cells add the same volume before cell division, meaning
that the relative growth rate of larger cells is slower or cell cycle is faster. Note that in both ‘sizer’ and ‘adder’ models the time to reach the critical cell cycle transition
may also vary, larger cells typically reaching G1/S or mitosis faster. (C) Size is jointly regulated by growth rate and cell cycle length. Here I use the expression by
Ginzberg et al. [16], but cell size could also be more generally expressed as a joint function of growth rate and cell cycle time (S = f(υ,τ)). Consequently, cell size can
be altered if growth rate or cell cycle length is altered. On the other hand, if cell size is constant, growth rate increase must be accompanied by decrease in cell cycle
length. (D) Kleiber's law indicates that metabolic activity is proportional to mass at organismal level. Provided that this is also true for individual cells, this formula
could potentially explain reduced growth rate of larger cells which is a requirement for robust cell size control in both ‘sizer’ and ‘adder’ models.

[27]. ‘Adder’ model postulates that both small and large cells add the from mean reversion as the daughter cells correct their size towards the
same volume during each cell cycle thus resulting in a stable cell size typical cell size. The size adjustment may take several cell divisions
distribution (Fig. 1B). The ‘adder’ model has been covered elsewhere in before the final target cell size is achieved. In contrast to the ‘sizer’
great detail [18,35], but the key point is that size homeostasis emerges model, the ‘adder’ model represents a more passive approach to

A B
CV of cell size
Cell count
Cell size

DNA content DNA content


Fig. 2. (A) Animal cells preferentially adjust their size at cell cycle transitions. Heatmap showing DNA content vs. cell size in Drosophila Kc167 cells shows reduction
in cell size variability at G1/S transition (white arrows). (B) The DNA content histogram depicting the cell cycle and median cell size variability (CV = coefficient of
variation; red dots) for the cells shown in (A). The cells were stained with Nuclear-ID Red DNA binding dye and analysed by flow cytometry. Cell size was measured
as forward scatter (FSC-A). The cell cycle stages G1, S and G2/M are shown along subG1 fraction (apoptosis/dead cell fraction).

411
M. Björklund BBA - Molecular Cell Research 1866 (2019) 409–417

maintain cell size homeostasis, as there is no acute requirement for cellular processes influencing cell size [40–42], although it has been
active size monitoring. challenging to identify which mechanisms are direct. For example, a
large number of cyclin dependent kinases, their inhibitors and activa-
3.2. ‘Sizer’ and ‘adder’ – more similarities than differences? tors regulate normal cell cycle progression and many of these cell cycle
proteins influence cell size [42]. It is also well established that animal
Despite the key conceptual differences in these models, there are cells can regulate their growth through multiple signaling pathways,
many similarities supported by the increasingly accurate experimental the PI3K-AKT-mTOR and RAS-MAPK pathways being the best char-
measurements. For example, both the ‘adder’ and ‘sizer’ models agree acterized examples [43].
that cell size homeostasis is maintained by joint regulation of growth Early work on bacterial growth established a relationship between
rate and cell cycle duration [16,23]. This is exemplified by a mathe- growth rate and cell size in a quantitative manner [44]. Essentially,
matical representation of how size relates to growth rate and cell cycle these analyses showed that faster growth rate in a richer culture
duration. Ginzberg et al. [16] proposed that size is simply the product medium results in larger cell size, establishing a connection between
of growth rate and cell cycle length (Fig. 1C). To maintain constant size, nutrient uptake, metabolic activity and cell size. While this is an in-
one cannot modify growth rate without changing cell cycle length and tuitive finding, it is much less clear how differences in cell size among
vice versa. This is consistent with common observation that cell cycle different cell types are achieved under identical culture conditions,
length in G1 is inversely proportional to cell size, i.e., small cells spend particularly in animal cells. By inference, such size differences could
more time in G1 before they start DNA replication. This equation may also be related to differences in nutrient uptake and metabolic effi-
equally well be expanded to relate to the ‘adder’ model as size in this ciency in converting these nutrients to biomass. A large body of ex-
equation could be either the constant target size (as in the ‘sizer’ model) periments in single cell organisms has established that growth rate and
or the constant volume added per cell cycle (as in the ‘adder’ model) nutrient availability are the primary determinants of cell size and also
(Fig. 1C). This simple mathematical representation how cell cycle influence cell cycle timing. Some of the mechanisms on how specific
length, growth rate and cell size are interconnected will hopefully help nutrients such as glucose act have been identified. For example, cell
to link the ‘adder’ and ‘sizer’ models with the molecular mechanisms division rate in budding yeast appears to depend on glucose influx
responsible for growth and cell cycle regulation, including metabolism while volume growth is largely set by external glucose levels [45].
and CDK regulation (see chapter 4). Other experiments point out that the nutrients (carbon source) have
Further similarities between the ‘adder’ and the ‘sizer’ models sug- clear effects on both duration of mitosis and cell size in yeast [46].
gest that these are not mutually exclusive, but rather different inter- Similarly, work in bacteria has identified specific nutrients such as
pretations of the same underlying biological mechanisms. Varsano et al. uridine diphosphate glucose (UDP-glucose) [47] or metabolic enzymes
reported that cultured mammalian cells that are roughly average size such as NAD-dependent glutamate dehydrogenase [48] as key meta-
display ‘adder’-like growth pattern, whereas abnormally small cells bolic coordinators of cell size and cell division. Furthermore, genetic
display growth pattern that requires reaching a certain size before evidence links nitrogen metabolism and pyruvate production to bac-
transition to S phase, consistent with ‘sizer’ model [26]. Similar findings terial cell division, although the molecular mechanisms are yet to be
have been reported for E. coli growth [36] and also suggested to apply determined [49].
to budding yeast [37]. Specifically, Wallden et al. found that DNA re- In contrast to these findings from unicellular organisms, there is
plication initiations in E. coli occur at integer multiples of a fixed vo- paucity of information regarding the role of specific nutrients and
lume, thus invalidating pure ‘timer’ and ‘adder’ models. They further metabolic enzymes in animal growth control. Most of our knowledge is
proposed a model based on timing of DNA replication initiation to ex- derived from experiments involving growth factor and metabolic sig-
plain this mode of growth where ‘adder’ becomes ‘sizer’ at slow growth naling pathways rather than the nutrients themselves, which may be
[36]. On the other hand, the model by Delarue et al. explains this two- related to the more complex nutritional requirements of animal cells. In
tier behavior by titration of an activator protein [37]. Overall these proliferating animal cells growth factors and growth factor signaling
results indicate that if the growth rate of the cells is sufficient, the (still pathways are major regulators of nutrient uptake and metabolic ac-
elusive) cell size checkpoints are not particularly strict and allow cells tivity, and these pathways also influence cell cycle progression [43].
of certain size range rather than a specific predetermined size to pass Although these growth factor signaling pathways have multiple out-
through these inspections points. puts, regulation of metabolism to support cellular biosynthesis is re-
Yet another shared feature of both the ‘adder’ and the ‘sizer’ models quired to achieve cell growth [43,50] (Fig. 3).
is that the robustness of size control in these models is dependent on the The mechanistic target of rapamycin (mTOR) is a good example
cell's ability to modulate growth rate and/or cell cycle progression in a illustrating the multifaceted role of signaling pathways in modulating
cell-size dependent manner. Even when cells add a constant volume cell growth and proliferation. The mTOR pathway is a key integrator of
each cell cycle as in the ‘adder’ model, the growth rate is not constant cellular growth and metabolism with external inputs such as nutrients
but is reduced in larger cells [23]. Currently the reason for such a size and growth factors [51]. This pathway appears to influence virtually
dependent growth rate can only be speculated. Kleiber's law states that every aspect of metabolism and has been directly linked to cell growth
the relative metabolic activity is reduced when organisms increase in and metabolism primarily via the mTORC1 complex as well as pro-
size (Fig. 1D). As metabolic activity is proportional to growth rate and liferation primarily via the mTORC2 complex [51]. However, this dis-
theoretical work suggests that metabolic allometry, the relationship tinction is not necessarily so clear-cut as for example recent work in
between metabolism and size, is universal across all scales of life [38], budding yeast indicates that both cell size and growth rate are modu-
this is potentially a useful framework for investigating size-dependent lated via mTORC2 complex [52]. Manipulation of mTOR activity in-
growth of cells [8]. Notably, such reduced growth in larger cells con- fluences animal cell size in many model systems [53,54]. Despite its
flicts with the purely exponential growth model (larger cells grow many roles in metabolic regulation, experiments with rapamycin and
faster), but is supported by experimental observations [7,16,23,39]. other mTOR inhibitors indicate that this kinase is not directly involved
in cell-size sensing [16] or setting the cell size dependent mitochondrial
4. From models to molecules and mechanisms: metabolic activity [7]. Therefore, despite the extensive understanding of the
regulation of cell size molecular details on mTOR signaling pathway [51], its true contribu-
tion to cell size regulation remains unresolved.
4.1. Growth signaling converges at metabolism The central carbon metabolism where sugar is converted to meta-
bolites and energy via glycolysis and tricarboxylic acid cycle in mi-
Genome-scale screens have identified many individual genes and tochondria forms the essential core for cellular metabolic networks and

412
M. Björklund BBA - Molecular Cell Research 1866 (2019) 409–417

Fig. 3. (A) Cells use nutrients such as glucose, glutamine and


A other amino acids as building blocks and also secrete waste
Glucose metabolites such as lactate and glutamate. Utilization of
nutrients by glycolysis, glutaminolysis and mitochondrial
Amino acids TCA cycle leads to generation of a wide variety of metabo-
lites for biosynthetic needs along energy (ATP) and redox
cofactors such as NADH and NADPH to support growth and
Lactate proliferation. In animal cells, metabolic activity is strongly
influenced by growth factors and the intracellular signaling
pathways such as PI3K-AKT-mTOR and RAS-MAPK path-
ways. (B) While glucose and glutamine are by far the most
Glutamate consumed nutrients, the carbon from these molecules con-
TCA CYCLE tributes relatively little to cellular biomass. Instead, amino
Glutamine acids other than glutamine make the biggest contribution to
cell mass [55]. The source of the remaining ~40% of carbon
has not been reliably identified. Part of this is derived from
lipids, especially palmitate and oleate [55].

e.g., mTORC1 e.g., mTORC2

Growth Proliferation

B
Glucose Glutamine Amino acids

Carbon up to up to up to
contribution ~10% ~10% ~40%
to biomass

is also essential for cell growth [43,50]. However, while the sugar-de- 4.2. Size-scaling of cell cycle regulators and metabolism
rived metabolites can be used to make biomass, experimental data from
cultured mammalian cells indicates that the majority of cell mass de- As discussed earlier, cell-size dependent growth rate regulation is
rives from amino acids rather than sugars [55] (Fig. 3B). Further important for cell size homeostasis. Size-scaling of macromolecule
complexity in the system arises from the findings that metabolite up- synthesis and/or metabolism could in part explain the relative reduc-
take is co-operative as in the presence of limiting amounts of a single tion of growth rate in larger cells. Overall, mRNA transcript abundance
nutrient (e.g., glucose), cells immediately adjust the uptake of other increases with cell size due to increased global transcription in larger
nutrients [56]. Understanding these complex metabolic requirements cells [11,12]. Similarly, the proteome scales with cell volume [13] and
and interdependencies for growth and cell division is likely to be es- displays a uniform scaling across different subcellular compartments
sential for developing effective therapeutic measures in cancers. It is and organelles across the cell cycle [7]. While overall macromolecular
well established that many cancer cell types increase glucose and glu- biosynthesis appears to follow cell size changes closely, there are in-
tamine uptake and reduce their dependency on mitochondrial oxidative dividual mRNAs and proteins that deviate from this general trend. Of
metabolism. The widespread use of this metabolic rewiring suggests particular interest are cell cycle activators and inhibitors, which could
that this provides a growth advantage not only to cancer cells but also couple cell size to cell cycle progression. While the general principle for
to other highly proliferative cell types, including activated lympho- the accumulation a critical amount of a specific protein was proposed
cytes, thymocytes and embryonic cells. already 40 years ago [31,61], such proteins are sought after even today.
Cellular metabolic activity and mitochondria in particular, not only For example, cell size could influence cell cycle transitions if size-
directly influences growth rate but also has an impact on cell cycle scaling of an inhibitor and activator are different. In budding yeast, the
progression through CDK and E2F activity [57]. The impact of meta- G1 cyclin Cln3 displays uniform size scaling [62]. In contrast, the levels
bolism on proliferation has been demonstrated with several in vivo of Cln3 inhibitor Whi5 are diluted when cells grow in G1 phase. This
models, including stem cells in Drosophila [58] and hepatocyte re- results in a threshold cell size where Whi5 levels cannot any more in-
generation in metabolic disorders [59,60]. Thus, metabolism is highly hibit Cln3 activity allowing the yeast cells to progress in cell cycle [62].
interconnected with both growth and cell cycle progression and per- An alternative cell cycle protein scaling has been proposed for cultured
haps even the cause for the observed coupling between growth, cell Drosophila cells [63]. This model involves the mitotic kinase CDK1 and
cycle and cell size (Fig. 3A). its inhibitors, which are membrane bound Myt1 and nuclear Wee1. In
small cells, surface area and nuclear volume to cell volume ratios are

413
M. Björklund BBA - Molecular Cell Research 1866 (2019) 409–417

high, resulting in inhibitory activity towards Cdk1. When the cell cause local differences in cellular metabolism and signaling resulting in
grows, the cytoplasm grows faster than the nucleus, and surface to targeted growth at specific cellular locations. While specific examples
volume ratio decreases, resulting in weakened inhibition of CDK1 and are relatively scarce, such a mechanism could be important for example
mitotic progression [63]. Mitotic regulation by cell size dependent ex- for localized mTOR dependent activation of translation in axons [74].
pression of Cdc25 in fission yeast is yet another example how cells For a more comprehensive discussion on cell shape induced local SV
couple their size and/or growth rate to cell division [64]. As Cdc25 is ratio changes, see [73]. Importantly, local membrane growth could
the phosphatase opposing the activities of CDK1 inhibitory kinases result in further changes in local SV ratio further reinforcing the con-
Wee1 and Myt1, this points out to the various ways how the master cell clusion that the relationship between growth and size is bidirectional.
cycle regulators such as Cdk1 can be regulated through size-scaling of A logical conclusion based on the observed bidirectionality between
their inhibitors or activators. Based on these examples, it is not difficult growth rate and cell size is that cells can sense their size. The cell size
to envision that such differences in scaling of a cell cycle activator to dependent changes in metabolism, as observed for example in mouse
inhibitor could represent a general, and possibly highly redundant, hepatocytes in vivo [12], suggests that metabolism, and consequently
mechanism to ensure size-dependent passage through various stages of growth rate, can be adjusted based on cell size. More generally, any
the cell cycle. Such titration-based mechanism could also be achieved cellular behavior consistent with size dependent adjustment of growth
through gene expression regulation. For example, if expression of a rate and/or cellular functions could be interpreted as a consequence of
specific DNA binding factor increases with cell size, this essentially cell size sensing. What could be the mechanism(s) enabling cells to
results in titration against a constant DNA amount, which would then measure their size?
trigger transcription after a specific threshold point is achieved. An
example of such a mechanism is histone titration during midblastula 5. Mechanisms for (apparent) cell size sensing
transition in frog oocytes [65].
Apart from scaling of individual cell cycle proteins, multiple mem- I already mentioned that if cells are able to sense their size, this
bers of a particular signaling pathway and/or a specific cellular process could be an important mechanism for the maintenance of appropriate
could display differential scaling with cell size. We have previously growth rate and/or cell cycle progression. However, some people might
found that the relative contribution of mitochondrial activity in mouse not agree that cells sense their size as the term ‘sensing’ implies active
liver is reduced when hepatocytes grow in size [12]. As mitochondrial monitoring. Nevertheless, it is helpful to think of different ways how
activity declines, hepatocytes are increasingly utilizing glycolysis and cells could measure their overall size or even the size of organelles or
other alternative biosynthetic pathways. Additional experiments with other specific cellular compartments [75,76].
cultured cells (both primary and immortalized) showed that mi- Experiments in unicellular model organisms have identified puta-
tochondrial activity displays a nonlinear size-dependency [7], con- tive ‘rulers’ or ‘size sensors’, including Pom1 and Cdr2 kinases in fission
sistent with observations of variable growth rate within the cell cycle yeast [77–79] and FtsZ and DnaA proteins in bacteria (reviewed in
[16,20,23]. Mechanistically, the changes in mitochondrial activity were [28]). Definitive evidence is still lacking, partly because of the lack of a
found to be dependent on mitochondrial dynamics, the balance be- clear consensus how a cell size sensor is defined. For example, it was
tween mitochondrial fusion and fission [7]. As the connectivity of mi- proposed originally that Pom1, a cell polarity protein kinase, acts as a
tochondrial network influences metabolic output [66,67], we specu- dose-dependent inhibitor of mitotic entry and forms a gradient from the
lated that this could represent a mechanism to regulate cellular growth cell ends towards the middle, essentially acting as a sensor for cell
rate and consequently cell size [8]. Mitochondrial fusion and fission are length (Fig. 4A). To investigate if Pom1 was an actual size sensor, Wood
regulated via multiple proteins, including mitofusins, OPA1, Fis1 and and Nurse [15] used two criteria. First, deleting a gene involved in size
Drp1 opening up possibilities to investigate their activity in regulation sensing should result in greater variability in cell size. Second, with the
of cell size and growth rate. size sensor deleted, the large and small cells should return less effi-
ciently to normal size. Based on these two criteria, they disputed that
4.3. Growth rate and cell size – a two-way road Pom1 acts as a ruler to measure cell size [15].
Redundancy in biological systems, especially in those processes
While specific details how cells adjust their metabolism and growth which are important for cellular fitness and viability, may be the reason
rate to regulate size remain elusive, some general trends can be ob- why bona fide size sensors have been difficult to identify. In addition, as
served. Most importantly, causal relationships are difficult to establish with man-made biosensors, it is possible that a cell size sensor is
due to the multidirectional relationship between metabolism, signaling composed of multiple components, which are separately involved in
and the cell cycle [68–72]. For example, bacterial physiologists see that size measurement and the transmission of the physical information into
cell size results from growth rate regulation [36,44]. While it may be biochemical and molecular information. Thus, genetic loss-of-function
intuitive to claim that cell size is the product resulting from the reg- experiments may fail to identify such multi-modular cell size sensors or
ulation of growth rate and cell division, biophysical reasoning argues sensor genes, which are essential for viability.
against this causality. This is because larger cell size should constrain An additional mechanism for cell size sensing is via cell area or SV
nutrient uptake and waste metabolite disposal and influence growth ratio (Fig. 4A). Cell area has been proposed as an alternative me-
rate, thus reversing the causality. Large cell size therefore makes me- chanism to cell length measurement in fission yeast [79] and SV ratio in
tabolism less efficient due to increases diffusion times and reduction in bacteria and animal cells [12,80,81]. In case of bacteria, it was sug-
the cell surface area relative to cell volume (SV ratio; for a more ex- gested that availability of bacterial cell wall precursors impacts SV
tensive discussion, see [8]). This difference in SV ratio could be one of homeostasis and consequently cell size. Similar mechanisms may also
the reasons for allometric scaling at the cellular level. In support to this be operational in animal cells as the mevalonate pathway that produces
biophysical reasoning of size – growth rate causality, ecologists typi- cholesterol, strongly influences cell size [12,82].
cally perceive that metabolic activity is a function of size as indicated It was proposed that the relative difference between surface and
by Kleiber's law. An objective interpretation of all this is that growth volume growth rather than the SV ratio itself is the key determinant for
rate and cell size influence each other. size control [80,81]. This further suggests that for geometric size sen-
Interestingly, the influence of the SV ratio may extend beyond cell sing, cells do not need to have a simple geometrical shape such as a
size as cell shape induced local changes in SV ratio could influence sphere or a rod. As long as cells are able to monitor excess external
metabolic activity and result in localized growth. Cell shape related surface component accumulation via any biochemical feedback me-
differences in SV ratio are particularly evident in migrating cells as well chanism, SV ratio monitoring can be accomplished. Whether the actual
as neurons [73]. These shape-related local SV changes could potentially SV ratio or the differential growth of surface and volume is important

414
M. Björklund BBA - Molecular Cell Research 1866 (2019) 409–417

Fig. 4. (A) Cells may sense their size through multiple mechanism, including local or global balance between the concentration of cell cycle activators [a] and
inhibitors [i]. For example, when local threshold is achieved as in case of Pom1 [i] gradient mediated inhibition of Cdr2 [a] in fission yeast, local Cdr2 activation
induces cell size dependent mitotic entry. A global, cell volume-related balance between Cln3 [a] and Whi5 [i] in budding yeast results in G1/S transition.
Alternatively, cells may sense their surface-to-volume ratio by monitoring the excess production of cell wall (e.g., in bacteria or plants) and/or cell membrane
material, illustrated as m (such as cholesterol in animal cells). This requires that accumulation of excess membrane material results in feedback that reduces cellular
volume growth and is independent of cell shape. (B) Proposed architecture of a putative cell size sensor. Such a sensor could comprise of a metabolic module that
senses either the growth rate or physical dimensions through one or more metabolic parameters. The size sensor is assumed to continuously monitor the cell size,
growth rate or metabolic rate. The output module is likely a cell cycle related protein and would cause growth rate adjustment only at cell size transitions such as G1/
S or G2/M, at other times size-sensing would be non-productive.

could be potentially dissected using 3D micro niches where individual 6. Concluding remarks
cells fill a predetermined shape [14], thus allowing one to analyse the
effect of SV ratio on cellular functions in the absence of growth. Recent single cell studies show that animal cell growth during the
Because cellular growth rate is so closely associated with metabolic cell cycle is not purely linear or exponential, necessitating the future
changes, it would be logical for cells to have a metabolic size sensor development of better and more realistic models. While the technical
rather than a kinase-based ruler that measures cell length. Sensing the measurements are now increasingly more accurate, it appears that one
differences in surface and volume growth, as in bacteria cells could be of the main limitations to understand cell size control is the inter-
one of the easiest ways to achieve metabolic size sensing [80,81]. Ex- pretation of experimental results. This is exemplified by the extra-
perimental data indicates that size is adjusted more strongly at the cell polation of the observed linear growth in S phase [3,4] and also illu-
cycle transitions in animal cells [16], but also in plants where the shoot strated by the fact that more than one model can equally well fit the
apical meristem cell size is regulated at both major cell cycle check- same high quality data [23,80]. In addition, single cell studies have
points (G1/S and mitosis) [83]. However, what we do not know for sure highlighted the fact that despite substantial variability in growth rate at
is if the molecular networks involved in size sensing are acting con- single cell level, this translates into stable cell size distribution and a
tinuously or only triggered at these specific times during the cell cycle. more predictable average growth rate at the population level. Some-
If the size sensing system is composed of two or more modules, the what counterintuitively, comparison of the ‘adder’ and ‘sizer’ models
sensing and information transmission could be decoupled so that cells suggests that proper maintenance of size homeostasis requires that
would only adjust their size in relation to cell cycle transitions, despite larger cells grow slower than small cells in relative terms. How do cells
continuous size monitoring. sense their size to modulate their growth rate accordingly? The mole-
Based on this, a cell size sensor could be composed of a metabolic cular mechanisms by which cells regulate their size remain poorly un-
module responsible for sensing cellular geometry and/or biosynthesis derstood and are likely to be more complex than anticipated. We need
and a cell cycle trigger module as an output (Fig. 4B). Such a sensor to clarify how different signaling pathways contribute to cell size reg-
would therefore link cell size to cell cycle progression and explain why ulation, particularly through regulation of metabolism. For example,
size adjustment occurs primarily at cell cycle transitions even if size while manipulation of mTOR activity alters cell size, this kinase is not a
monitoring would be continuous. While the quest for the cell size sensor key contributor to cell-size sensing in animal cells [14]. Instead, the p38
is ongoing, it is important to remember that the apparent size-sensing MAPK pathway reduces cell size variability while coordinating cell size
could also be an emergent property of the system rather than active and cell cycle progression [33]. Neither is mTOR activity involved in
cellular computation based on the measurement of a single parameter setting the cell size dependent mitochondrial activity, whereas the
related to cellular dimensions. mevalonate/cholesterol pathway is involved [5]. How can these results
be reconciled? Does the whole question of cell size control need to be
subdivided into two or more processes? The role of CDK4 in regulating

415
M. Björklund BBA - Molecular Cell Research 1866 (2019) 409–417

cell's target size also warrants further investigation as this may directly [20] A. Tzur, R. Kafri, V.S. LeBleu, G. Lahav, M.W. Kirschner, Cell growth and size
link cell size regulation with cancer as specific CDK4 inhibitors have homeostasis in proliferating animal cells, Science 325 (2009) 167–171, https://doi.
org/10.1126/science.1174294.
remarkable activity particularly in breast cancer [84]. While many [21] E.A. Corbin, O.O. Adeniba, O.V. Cangellaris, W.P. King, R. Bashir, Evidence of
questions remain open, the recent advances clearly imply that we are differential mass change rates between human breast cancer cell lines in culture,
now several steps closer in understanding cell size control and the Biomed. Microdevices 19 (2017) 10, https://doi.org/10.1007/s10544-017-0151-x.
[22] N. Cermak, S. Olcum, F.F. Delgado, S.C. Wasserman, K.R. Payer, M. A Murakami,
implications of cell size homeostasis for human health. S.M. Knudsen, R.J. Kimmerling, M.M. Stevens, Y. Kikuchi, A. Sandikci, M. Ogawa,
V. Agache, F. Baléras, D.M. Weinstock, S.R. Manalis, High-throughput measurement
Conflict of interests of single-cell growth rates using serial microfluidic mass sensor arrays, Nat.
Biotechnol. 34 (2016) 1052–1059, https://doi.org/10.1038/nbt.3666.
[23] C. Cadart, S. Monnier, J. Grilli, P.J. Sáez, N. Srivastava, R. Attia, E. Terriac,
The author declares no conflict of interests. B. Baum, M. Cosentino-Lagomarsino, M. Piel, Size control in mammalian cells in-
volves modulation of both growth rate and cell cycle duration, Nat. Commun. 9
(2018) 3275, https://doi.org/10.1038/s41467-018-05393-0.
Transparency document
[24] A. Sveiczer, B. Novak, J.M. Mitchison, The size control of fission yeast revisited, J.
Cell Sci. 2957 (1996) 2947–2957.
The Transparency document associated with this article can be [25] A.I. Goranov, M. Cook, M. Ricicova, G. Ben-Ari, C. Gonzalez, C. Hansen, M. Tyers,
found, in online version. A. Amon, The rate of cell growth is governed by cell cycle stage, Genes Dev. 23
(2009) 1408–1422, https://doi.org/10.1101/gad.1777309.
[26] G. Varsano, Y. Wang, M. Wu, Probing mammalian cell size homeostasis by channel-
Acknowledgements assisted cell reshaping, Cell Rep. 20 (2017) 397–410, https://doi.org/10.1016/j.
celrep.2017.06.057.
[27] N.P. Fantes P, Division timing: controls, models and mechanisms, in: P.C.L. John
The author wants to thank Dr. T. Miettinen and Dr. K.Y. Chan for (Ed.), Cell Cycle, Cambridge University Press, London, 1981.
critical reading of the manuscript. This work was supported by funds [28] L. Robert, Size sensors in bacteria, cell cycle control, and size control, Front.
from Zhejiang University. Microbiol. 6 (2015) 515, https://doi.org/10.3389/fmicb.2015.00515.
[29] H. Dolznig, F. Grebien, T. Sauer, H. Beug, E.W. Müllner, Evidence for a size-sensing
mechanism in animal cells, Nat. Cell Biol. 6 (2004) 899–905, https://doi.org/10.
References 1038/ncb1166.
[30] A.J. Kerkhoff, B.J. Enquist, Multiplicative by nature: why logarithmic transforma-
tion is necessary in allometry, J. Theor. Biol. 257 (2009) 519–521, https://doi.org/
[1] A.C. Lloyd, The regulation of cell size, Cell 154 (2013) 1194–1205, https://doi.org/
10.1016/j.jtbi.2008.12.026.
10.1016/j.cell.2013.08.053.
[31] P.W. Rossow, V.G. Riddle, A.B. Pardee, Synthesis of labile, serum-dependent protein
[2] M.B. Ginzberg, R. Kafri, M. Kirschner, On being the right (cell) size, Science 348
in early G1 controls animal cell growth, Proc. Natl. Acad. Sci. U. S. A. 76 (1979)
(2015) 1245075, https://doi.org/10.1126/science.1245075.
4446–4450, https://doi.org/10.1073/pnas.76.9.4446.
[3] I.J. Conlon, G.A. Dunn, A.W. Mudge, M.C. Raff, Extracellular control of cell size,
[32] P. Dong, C. Zhang, B.T. Parker, L. You, B. Mathey-Prevot, Cyclin D/CDK4/6 activity
Nat. Cell Biol. 3 (2001) 918–921, https://doi.org/10.1038/ncb1001-918.
controls G1 length in mammalian cells, PLoS One 13 (2018) e0185637, , https://
[4] I. Conlon, M. Raff, Differences in the way a mammalian cell and yeast cells co-
doi.org/10.1371/journal.pone.0185637.
ordinate cell growth and cell-cycle progression, J. Biol. 2 (2003) 7.
[33] S. Liu, M.B. Ginzberg, N. Patel, M. Hild, B. Leung, Z. Li, Y.C. Chen, N. Chang,
[5] A. Amir, Is cell size a spandrel? elife 6 (2017), https://doi.org/10.7554/eLife.
Y. Wang, C. Tan, S. Diena, W. Trimble, L. Wasserman, J.L. Jenkins, M.W. Kirschner,
22186.
R. Kafri, Size uniformity of animal cells is actively maintained by a p38 MAPK-
[6] J.T. Bonner, Why Size Matters: From Bacteria to Blue Whales, Princeton University
dependent regulation of G1-length, elife 7 (2018), https://doi.org/10.7554/eLife.
Press, 2006.
26947.
[7] T.P. Miettinen, M. Björklund, Cellular allometry of mitochondrial functionality
[34] O. Casanovas, F. Miró, J.M. Estanyol, E. Itarte, N. Agell, O. Bachs, Osmotic stress
establishes the optimal cell size, Dev. Cell 39 (2016) 370–382, https://doi.org/10.
regulates the stability of cyclin D1 in a p38SAPK2-dependent manner, J. Biol.
1016/j.devcel.2016.09.004.
Chem. 275 (2000) 35091–35097, https://doi.org/10.1074/jbc.M006324200.
[8] T.P. Miettinen, M. Björklund, Mitochondrial function and cell size: an allometric
[35] J.T. Sauls, D. Li, S. Jun, Adder and a coarse-grained approach to cell size home-
relationship, Trends Cell Biol. 27 (2017) 393–402, https://doi.org/10.1016/j.tcb.
ostasis in bacteria, Curr. Opin. Cell Biol. 38 (2016) 38–44, https://doi.org/10.
2017.02.006.
1016/j.ceb.2016.02.004.
[9] J.A. Vernon, W.H. Frank, W.V. Slack, Effect of size and number of brain cells on
[36] M. Wallden, D. Fange, E.G. Lundius, Ö. Baltekin, J. Elf, The synchronization of
learning in larvae of the salamander, Triturus viridescens, Science 122 (1955)
replication and division cycles in individual E. coli cells, Cell 166 (2016) 729–739,
692–693.
https://doi.org/10.1016/j.cell.2016.06.052.
[10] S. Anzi, M. Stolovich-Rain, A. Klochendler, O. Fridlich, A. Helman, A. Paz-
[37] M. Delarue, D. Weissman, O. Hallatschek, A simple molecular mechanism explains
Sonnenfeld, N. Avni-Magen, E. Kaufman, M.B. Ginzberg, D. Snider, S. Ray,
multiple patterns of cell-size regulation, PLoS One 12 (2017) e0182633, , https://
M. Brecht, M.M. Holmes, K. Meir, A. Avivi, I. Shams, A. Berkowitz, A.M.J. Shapiro,
doi.org/10.1371/journal.pone.0182633.
B. Glaser, S. Ben-Sasson, R. Kafri, Y. Dor, Postnatal exocrine pancreas growth by
[38] G.B. West, W.H. Woodruff, J.H. Brown, Allometric scaling of metabolic rate from
cellular hypertrophy correlates with a shorter lifespan in mammals, Dev. Cell 45
molecules and mitochondria to cells and mammals, Proc. Natl. Acad. Sci. 99 (2002)
(2018) 726–737.e3, https://doi.org/10.1016/j.devcel.2018.05.024.
2473–2478, https://doi.org/10.1073/pnas.012579799.
[11] O. Padovan-Merhar, G.P. Nair, A.G. Biaesch, A. Mayer, S. Scarfone, S.W. Foley,
[39] K. Park, L.J. Millet, N. Kim, H. Li, X. Jin, G. Popescu, N.R. Aluru, K.J. Hsia,
A.R. Wu, L.S. Churchman, A. Singh, A. Raj, Single mammalian cells compensate for
R. Bashir, Measurement of adherent cell mass and growth, Proc. Natl. Acad. Sci. 107
differences in cellular volume and DNA copy number through independent global
(2010) 20691–20696, https://doi.org/10.1073/pnas.1011365107.
transcriptional mechanisms, Mol. Cell 58 (2015) 339–352, https://doi.org/10.
[40] R. Kittler, L. Pelletier, A.K. Heninger, M. Slabicki, M. Theis, L. Miroslaw, I. Poser,
1016/j.molcel.2015.03.005.
S. Lawo, H. Grabner, K. Kozak, J. Wagner, V. Surendranath, C. Richter, W. Bowen,
[12] T.P. Miettinen, H.K.J. Pessa, M.J. Caldez, T. Fuhrer, M.K. Diril, U. Sauer, P. Kaldis,
A.L. Jackson, B. Habermann, A.A. Hyman, F. Buchholz, Genome-scale RNAi pro-
M. Björklund, Identification of transcriptional and metabolic programs related to
filing of cell division in human tissue culture cells, Nat. Cell Biol. 9 (2007)
mammalian cell size, Curr. Biol. 24 (2014) 598–608, https://doi.org/10.1016/j.
1401–1412, https://doi.org/10.1038/ncb1659.
cub.2014.01.071.
[41] P. Jorgensen, J.L. Nishikawa, B.J. Breitkreutz, M. Tyers, Systematic identification of
[13] S. Marguerat, A. Schmidt, S. Codlin, W. Chen, R. Aebersold, J. Bähler, Quantitative
pathways that couple cell growth and division in yeast, Science 297 (2002)
analysis of fission yeast transcriptomes and proteomes in proliferating and quies-
395–400, https://doi.org/10.1126/science.1070850.
cent cells, Cell 151 (2012) 671–683, https://doi.org/10.1016/j.cell.2012.09.019.
[42] M. Björklund, M. Taipale, M. Varjosalo, J. Saharinen, J. Lahdenperä, J. Taipale,
[14] M. Bao, J. Xie, A. Piruska, W.T.S. Huck, 3D microniches reveal the importance of
Identification of pathways regulating cell size and cell-cycle progression by RNAi,
cell size and shape, Nat. Commun. 8 (2017) 1962, https://doi.org/10.1038/
Nature 439 (2006) 1009–1013, https://doi.org/10.1038/nature04469.
s41467-017-02163-2.
[43] W. Palm, C.B. Thompson, Nutrient acquisition strategies of mammalian cells,
[15] E. Wood, P. Nurse, Pom1 and cell size homeostasis in fission yeast, Cell Cycle 12
Nature 546 (2017) 234–242, https://doi.org/10.1038/nature22379.
(2013) 3228–3236, https://doi.org/10.4161/cc.26462.
[44] M. Schaechter, O. Maaloe, N.O. Kjeldgaard, Dependency on medium and tem-
[16] M.B. Ginzberg, N. Chang, H. D'Souza, N. Patel, R. Kafri, M.W. Kirschner, Cell size
perature of cell size and chemical composition during balanced growth of
sensing in animal cells coordinates anabolic growth rates and cell cycle progression
Salmonella typhimurium, J. Gen. Microbiol. 19 (1958) 592–606, https://doi.org/
to maintain cell size uniformity, elife 7 (2018), https://doi.org/10.7554/eLife.
10.1099/00221287-19-3-592.
26957.
[45] H. Schmidt-Glenewinkel, N. Barkai, Loss of growth homeostasis by genetic decou-
[17] S. Jun, S. Taheri-Araghi, Cell-size maintenance: universal strategy revealed, Trends
pling of cell division from biomass growth: implication for size control mechanisms,
Microbiol. 23 (2015) 4–6, https://doi.org/10.1016/j.tim.2014.12.001.
Mol. Syst. Biol. 10 (2014) 769.
[18] K.M. Schmoller, The phenomenology of cell size control, Curr. Opin. Cell Biol. 49
[46] R.M. Leitao, D.R. Kellogg, The duration of mitosis and daughter cell size are
(2017) 53–58, https://doi.org/10.1016/j.ceb.2017.11.011.
modulated by nutrients in budding yeast, J. Cell Biol. 216 (2017) 3463–3470,
[19] S.S. Grewal, B.A. Edgar, Controlling cell division in yeast and animals: does size
https://doi.org/10.1083/jcb.201609114.
matter? J. Biol. 2 (2003) 5.
[47] N.S. Hill, P.J. Buske, Y. Shi, P.A. Levin, A moonlighting enzyme links Escherichia coli

416
M. Björklund BBA - Molecular Cell Research 1866 (2019) 409–417

cell size with central metabolism, PLoS Genet. 9 (2013) e1003663, , https://doi. pnas.1413990112.
org/10.1371/journal.pgen.1003663. [66] M. Liesa, O.S. Shirihai, Mitochondrial dynamics in the regulation of nutrient utili-
[48] F. Beaufay, J. Coppine, A. Mayard, G. Laloux, X. De Bolle, R. Hallez, A NAD-de- zation and energy expenditure, Cell Metab. 17 (2013) 491–506, https://doi.org/10.
pendent glutamate dehydrogenase coordinates metabolism with cell division in 1016/j.cmet.2013.03.002.
Caulobacter crescentus, EMBO J. 34 (2015) 1786–1800, https://doi.org/10.15252/ [67] E. Schrepfer, L. Scorrano, Mitofusins, from mitochondria to metabolism, Mol. Cell
embj.201490730. 61 (2016) 683–694, https://doi.org/10.1016/j.molcel.2016.02.022.
[49] C.S. Westfall, P.A. Levin, Bacterial cell size: multifactorial and multifaceted, Annu. [68] C.G. Havens, A. Ho, N. Yoshioka, S.F. Dowdy, Regulation of late G1/S phase
Rev. Microbiol. 71 (2017) 499–517, https://doi.org/10.1146/annurev-micro- transition and APCCdh1 by reactive oxygen species, Mol. Cell. Biol. 26 (2006)
090816-093803. 4701–4711, https://doi.org/10.1128/MCB.00303-06.
[50] J.W. Locasale, L.C. Cantley, Metabolic flux and the regulation of mammalian cell [69] S.G. Menon, E.H. Sarsour, D.R. Spitz, R. Higashikubo, M. Sturm, H. Zhang,
growth, Cell Metab. 14 (2011) 443–451, https://doi.org/10.1016/j.cmet.2011.07. P.C. Goswami, Redox regulation of the G1 to S phase transition in the mouse em-
014. bryo fibroblast cell cycle, Cancer Res. 63 (2003) 2109–2117.
[51] R.A. Saxton, D.M. Sabatini, mTOR signaling in growth, metabolism, and disease, [70] K.E. Wellen, C.B. Thompson, A two-way street: reciprocal regulation of metabolism
Cell 168 (2017) 960–976, https://doi.org/10.1016/j.cell.2017.02.004. and signalling, Nat. Rev. Mol. Cell Biol. 13 (2012) 270–276, https://doi.org/10.
[52] R. Lucena, M. Alcaide-Gavilán, K. Schubert, M. He, M.G. Domnauer, C. Marquer, 1038/nrm3305.
C. Klose, M.A. Surma, D.R. Kellogg, Cell size and growth rate are modulated by [71] A.M. Ambrus, A.B.M.M.K. Islam, K.B. Holmes, N. Moon, N. Lopez-Bigas,
TORC2-dependent signals, Curr. Biol. 28 (2018) 196–210.e4, https://doi.org/10. E.V. Benevolenskaya, M.V. Frolov, Loss of dE2F compromises mitochondrial func-
1016/j.cub.2017.11.069. tion, Dev. Cell 27 (2013) 438–451, https://doi.org/10.1016/j.devcel.2013.10.002.
[53] D.C. Fingar, S. Salama, C. Tsou, E. Harlow, J. Blenis, Mammalian cell size is con- [72] J.A. Siqueira, P. Hardoim, P.C.G. Ferreira, A. Nunes-Nesi, A.S. Hemerly, Unraveling
trolled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E, Genes Dev. 16 interfaces between energy metabolism and cell cycle in plants, Trends Plant Sci.
(2002) 1472–1487, https://doi.org/10.1101/gad.995802. (2018), https://doi.org/10.1016/j.tplants.2018.05.005.
[54] H. Zhang, J.P. Stallock, J.C. Ng, C. Reinhard, T.P. Neufeld, Regulation of cellular [73] A. Haupt, N. Minc, How cells sense their own shape – mechanisms to probe cell
growth by the Drosophila target of rapamycin dTOR, Genes Dev. 14 (2000) geometry and their implications in cellular organization and function, J. Cell Sci.
2712–2724, https://doi.org/10.1101/gad.835000. 131 (2018) jcs214015, , https://doi.org/10.1242/jcs.214015.
[55] A.M. Hosios, V.C. Hecht, L.V. Danai, M.O. Johnson, J.C. Rathmell, M.L. Steinhauser, [74] M. Terenzio, G. Schiavo, M. Fainzilber, Compartmentalized signaling in neurons:
S.R. Manalis, M.G. Vander Heiden, Amino acids rather than glucose account for the from cell biology to neuroscience, Neuron 96 (2017) 667–679, https://doi.org/10.
majority of cell mass in proliferating mammalian cells, Dev. Cell 36 (2016) 1016/j.neuron.2017.10.015.
540–549, https://doi.org/10.1016/j.devcel.2016.02.012. [75] W.F. Marshall, Cell geometry: how cells count and measure size, Annu. Rev.
[56] S. Son, M.M. Stevens, H.X. Chao, C. Thoreen, A.M. Hosios, L.D. Schweitzer, Biophys. 45 (2016) 49–64, https://doi.org/10.1146/annurev-biophys-062215-
Y. Weng, K. Wood, D. Sabatini, M.G. Vander Heiden, S. Manalis, Cooperative nu- 010905.
trient accumulation sustains growth of mammalian cells, Sci. Rep. 5 (2015) 17401, [76] P. Jorgensen, M. Tyers, How cells coordinate growth and division, Curr. Biol. 14
https://doi.org/10.1038/srep17401. (2004) R1014–R1027, https://doi.org/10.1016/j.cub.2004.11.027.
[57] V. Fritz, L. Fajas, Metabolism and proliferation share common regulatory pathways [77] S.G. Martin, M. Berthelot-Grosjean, Polar gradients of the DYRK-family kinase
in cancer cells, Oncogene 29 (2010) 4369–4377, https://doi.org/10.1038/onc. Pom1 couple cell length with the cell cycle, Nature 459 (2009) 852–856, https://
2010.182. doi.org/10.1038/nature08054.
[58] C.C.F. Homem, V. Steinmann, T.R. Burkard, A. Jais, H. Esterbauer, J.A. Knoblich, [78] J.B. Moseley, A. Mayeux, A. Paoletti, P. Nurse, A spatial gradient coordinates cell
Ecdysone and mediator change energy metabolism to terminate proliferation in size and mitotic entry in fission yeast, Nature 459 (2009) 857–860, https://doi.org/
drosophila neural stem cells, Cell 158 (2014) 874–888, https://doi.org/10.1016/j. 10.1038/nature08074.
cell.2014.06.024. [79] K.Z. Pan, T.E. Saunders, I. Flor-Parra, M. Howard, F. Chang, Cortical regulation of
[59] R.A. DeAngelis, M.M. Markiewski, R. Taub, J.D. Lambris, A high-fat diet impairs cell size by a sizer cdr2p, elife 3 (2014) e02040, , https://doi.org/10.7554/eLife.
liver regeneration in C57BL/6 mice through overexpression of the NF-κB inhibitor, 02040.
IκBα, Hepatology 42 (2005) 1148–1157, https://doi.org/10.1002/hep.20879. [80] L.K. Harris, J.A. Theriot, Surface area to volume ratio: a natural variable for bac-
[60] R. Veteläinen, A.K. Van Vliet, T.M. Van Gulik, Severe steatosis increases hepato- terial morphogenesis, Trends Microbiol. (2018), https://doi.org/10.1016/j.tim.
cellular injury and impairs liver regeneration in a rat model of partial hepatectomy, 2018.04.008.
Ann. Surg. 245 (2007) 44–50, https://doi.org/10.1097/01.sla.0000225253. [81] L.K. Harris, J.A. Theriot, Relative rates of surface and volume synthesis set bacterial
84501.0e. cell size, Cell 165 (2016) 1479–1492, https://doi.org/10.1016/j.cell.2016.05.045.
[61] J. Campisi, E.E. Medrano, G. Morreo, A.B. Pardee, Restriction point control of cell [82] T.P. Miettinen, M. Björklund, Mevalonate pathway regulates cell size homeostasis
growth by a labile protein: evidence for increased stability in transformed cells, and Proteostasis through autophagy, Cell Rep. 13 (2015) 2610–2620, https://doi.
Proc. Natl. Acad. Sci. U. S. A. 79 (1982) 436–440. org/10.1016/j.celrep.2015.11.045.
[62] K.M. Schmoller, J.J. Turner, M. Kõivomägi, J.M. Skotheim, Dilution of the cell cycle [83] A. R. Jones, M. Forero-Vargas, S.P. Withers, R.S. Smith, J. Traas, W. Dewitte,
inhibitor Whi5 controls budding-yeast cell size, Nature 526 (2015) 268–272, J.A.H. Murray, Cell-size dependent progression of the cell cycle creates homeostasis
https://doi.org/10.1038/nature14908. and flexibility of plant cell size, Nat. Commun. 8 (2017) 15060, https://doi.org/10.
[63] M. Bonke, M. Turunen, M. Sokolova, A. Vähärautio, T. Kivioja, M. Taipale, 1038/ncomms15060.
M. Björklund, J. Taipale, Transcriptional networks controlling the cell cycle, G3 [84] M. Cristofanilli, N.C. Turner, I. Bondarenko, J. Ro, S.-A. Im, N. Masuda, M. Colleoni,
(Bethesda) 3 (2013) 75–90, https://doi.org/10.1534/g3.112.004283. A. DeMichele, S. Loi, S. Verma, H. Iwata, N. Harbeck, K. Zhang, K.P. Theall,
[64] D. Keifenheim, X.-M. Sun, E. D'Souza, M.J. Ohira, M. Magner, M.B. Mayhew, Y. Jiang, C.H. Bartlett, M. Koehler, D. Slamon, Fulvestrant plus palbociclib versus
S. Marguerat, N. Rhind, Size-dependent expression of the mitotic activator Cdc25 fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative
suggests a mechanism of size control in fission yeast, Curr. Biol. 27 (2017) metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-
1491–1497.e4, https://doi.org/10.1016/j.cub.2017.04.016. 3): final analysis of the multicentre, double-blind, phase 3 randomised controlled
[65] A.A. Amodeo, D. Jukam, A.F. Straight, J.M. Skotheim, Histone titration against the trial, Lancet Oncol. 17 (2016) 425–439, https://doi.org/10.1016/S1470-2045(15)
genome sets the DNA-to-cytoplasm threshold for the Xenopus midblastula transition, 00613-0.
Proc. Natl. Acad. Sci. U. S. A. 112 (2015) E1086–E1095, https://doi.org/10.1073/

417

S-ar putea să vă placă și