You are on page 1of 8

Cell Calcium 70 (2018) 16–23

Contents lists available at ScienceDirect

Cell Calcium
journal homepage: www.elsevier.com/locate/ceca

Review

Calcium signaling and cellular senescence


Nadine Martin a,b,c,d , David Bernard a,b,c,d,∗
a
Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69373 Lyon, France
b
CNRS UMR 5286, F-69373 Lyon, France
c
Centre Léon Bérard, F-69373 Lyon, France
d
Université de Lyon, F-69373 Lyon, France

a r t i c l e i n f o a b s t r a c t

Article history: Cellular senescence is a stable cell proliferation arrest induced by a variety of stresses including telom-
Received 28 February 2017 ere shortening, oncogene activation and oxidative stress. This process plays a crucial role in many
Received in revised form 3 April 2017 physiopathological contexts, especially during aging when cellular senescence favors development of
Accepted 3 April 2017
age-related diseases, shortening lifespan. However, the molecular and cellular mechanisms controlling
Available online 5 April 2017
senescence are still a matter of active research. In the last decade, there has been emerging literature indi-
cating a key involvement of calcium signaling in cellular senescence. In this review we will initially give
Keywords:
an account of the direct evidence linking calcium and the regulation of senescence. We will then review
Calcium
Senescence our current knowledge on the role of calcium in some senescence-associated features and physiopatho-
Signaling logical conditions, which will shed light on additional ways in which calcium signaling is implicated in
Cancer cellular senescence.
Aging © 2017 Elsevier Ltd. All rights reserved.

Contents

1. Definition of cellular senescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17


2. Direct evidence of the role of calcium in senescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
2.1. Increase in intracellular calcium levels in senescent cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
2.2. Calcium-induced mitochondrial dysfunction and ROS production in senescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
2.3. SASP regulation by calcium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
2.4. Role of the calcium/NFAT signaling pathway in senescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
3. Other roles for calcium in the main senescence-associated features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
3.1. Cell proliferation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
3.2. Senescence-associated secretory phenotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4. Role of calcium in senescence-associated physiopathological conditions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4.1. Role of cellular senescence in vivo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4.2. Implication of calcium in some physiopathological responses controlled by senescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
5. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

Abbreviations: 4EBP1, Eukaryotic Translation Initiation Factor 4E Binding Protein 1; ATF3, activating transcription factor 3; ATM, Ataxia Telangiectasia Mutated; ATR, Ataxia
Telangiectasia and Rad3-Related; ARF, alternative reading frame; BAPTA, 1,2-bis(o-aminophenoxy)ethane-N,N,N ,N -tetraacetic acid; CAMK, calmodulin kinase; CDK, cyclin
dependent kinase; CDKN1A/2A, cyclin dependent kinase inhibitor 1A/2A; CHK2, checkpoint 2 kinase; CEBP-␤, CCAAT/enhancer-binding protein beta; CRE, cAMP response ele-
ment; CREB, cAMP responsive element binding protein; DDR, DNA damage response; DUOX, dual oxidase 1; ER, endoplasmic reticulum; IL, interleukin; IKB␣, I-kappa-B alpha;
IKK, I-kappa-B kinase; IP3, inositol-1,4,5-trisphosphate; IP3R, inositol-1,4,5-trisphosphate receptor; mTOR, mammalian target of rapamycin; mTORC1, mammalian target of
rapamycin complex 1; NFAT, nuclear factor of activated T-cells; NF␬B, nuclear factor kappa-light-chain-enhancer of activated B cells; MAM, mitochondria-associated endo-
plasmic reticulum membranes; MCU, mitochondrial calcium uniporter; OIS, oncogene-induced senescence; PDGF, platelet-derived growth factor; PLC, phospholipase C; PKB,
protein kinase B; PML, promyelocytic leukemia protein; PPAR␥, peroxisome proliferator-activated receptor gamma; PTEN, phosphatase and tensin homolog; RB, retinoblas-
toma protein; RHEB1, ras homolog enriched in brain 1; ROS, reactive oxygen species; S6K, ribosomal protein S6 kinase; SAHF, senescence-associated heterochromatin foci;
SASP, senescence-associated secretory phenotype; TRP, transient receptor potential channel; TSC1/2, tuberous sclerosis 1/2.
∗ Corresponding author at: Centre de Recherche en Cancérologie de Lyon, F-69373 Lyon, France.
E-mail address: david.bernard@lyon.unicancer.fr (D. Bernard).

https://doi.org/10.1016/j.ceca.2017.04.001
0143-4160/© 2017 Elsevier Ltd. All rights reserved.

Descargado para Anonymous User (n/a) en University of Chile de ClinicalKey.es por Elsevier en febrero 04, 2021.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
N. Martin, D. Bernard / Cell Calcium 70 (2018) 16–23 17

1. Definition of cellular senescence membrane calcium channels or to calcium release from the endo-
plasmic reticulum (ER) depending on the context. Indeed, it has
Cellular senescence was originally described five decades ago as been shown that isradipine, a CaV1.3 L-type channel antagonist,
the proliferation arrest that accompanies the exhaustion of replica- prevents the rise in rotenone-induced calcium that accelerates
tive potential in primary human fibroblasts after extended culture senescence in human neuroblastoma SH-SY5Y cells [15]. In human
in vitro. Telomere attrition is the main trigger of this so-called mammary epithelial cells and primary human fibroblasts, onco-
replicative senescence. However, a number of other stimuli such as gene activation and telomere shortening trigger calcium release
oncogenic signals, oxidative stress and genotoxic agents can also from endoplasmic reticulum stores, through the activation of
induce a senescent phenotype, known as premature senescence. the PLC/IP3/IP3R pathway [16]. In H2 O2 -induced senescence in
Cellular senescence is thus generally defined as a cellular response human endometrium-derived stem cells, this IP3R-mediated cal-
to stresses. Although originally observed in primary human fibrob- cium release from the ER causes the rapid increase in cytosolic
lasts, cellular senescence has now been observed in most human calcium concentration [17]. Treatment with uridine triphosphate
cell types and in many species [1–4]. or thapsigargin, which deplete the ER calcium store, significantly
The two main features of senescent cells are their stable cell impaired the rise H2 O2 -induced intracellular calcium. High concen-
cycle arrest and their distinct, often pro-inflammatory, secretome trations of intracellular calcium are sustained during senescence.
or senescence-associated secretory phenotype (SASP). P53 and RB Chelation of this calcium with BAPTA [17] or knockdown of the ER
(retinoblastoma) are master regulators of senescence implement- calcium release channels IP3R [16] foster escape from senescence,
ing the stable cell cycle arrest. Upon stresses, p53 is activated and demonstrating that elevation of intracellular calcium levels triggers
induces the expression of genes such as CDKN1A encoding the cellular senescence.
cyclin-dependent kinase inhibitor p21. P21 and p16 (CDKN2A), Interestingly, several oncogenes and tumor suppressors regu-
another inhibitor of cyclin-dependent kinases, then inhibit the lating cellular senescence physically interact with IP3Rs and, by
phosphorylation of RB. RB consequently represses the expression modulating the activity of these channels, exploit calcium signal-
of pro-proliferative genes such as E2F target genes promoting the ing [18]. For example, it has been shown that IP3R3 is targeted by
cell cycle G1-S transition (Fig. 1) [5,6]. PKB/AKT pro-survival serine/threonine kinase [19]. This oncogene
The SASP comprises many pro-inflammatory cytokines and inhibits IP3R3-mediated ER calcium release by phosphorylating
chemokines, various growth factors and proteases. This secre- this channel [20]. The tumor suppressor PML (promyelocytic
tome is regulated both at the transcriptional level, the expression leukemia protein), a well know senescence inducer [21], forms
of numerous SASP components being induced by transcription a complex with IP3R3 and PKB/AKT and recruits the PP2A phos-
factors such as nuclear factor kappa-light-chain-enhancer of acti- phatase to counteract PKB/AKT-induced IP3R3 phosphorylation
vated B cells (NF␬B) and CCAAT/enhancer-binding protein beta and inhibition. The resulting increase in ER calcium release is
(CEBP-␤), and at the translational level by pathways such as instrumental in PML-induced apoptosis [22]. It can be hypothesized
the mammalian target of rapamycin (mTOR) pathway. The SASP that it may also be involved in PML-induced senescence.
displays autocrine activities reinforcing senescence as well as
several paracrine activities, sometime having opposite effects:
activating the immune system, spreading senescence and/or pro-
2.2. Calcium-induced mitochondrial dysfunction and ROS
moting epithelial-mesenchymal transition, migration and invasion
production in senescence
in neighboring cells (Fig. 1) [7–14].
Besides these two main features, senescent cells acquire other
Calcium released from the ER in response to senescence-
characteristics. They undergo changes in their morphology, which
inducing stresses mainly exerts its effects through reactive oxygen
often become flat and enlarged. They exhibit senescence-associated
species (ROS). It has been shown upon oncogene-induced senes-
␤-galactosidase activity, which is experimentally used as a marker
cence (OIS) in mammary epithelial cells and upon replicative
of senescence. They display increased levels of reactive oxygen
senescence in primary human fibroblasts that calcium release
species (ROS) which could result from altered mitochondrial activ-
from the ER is followed by its accumulation in the mitochondria.
ity and lead to DNA damage. They may also accumulate unrepaired
The increase in mitochondrial calcium concentration leads to a
DNA damages, which activate the ATM/ATR-p53-p21-RB DNA dam-
drop in mitochondrial membrane potential and in the enhanced
age response (DDR) pathway, as well as senescence-associated
production of ROS, which triggers senescence. Knockdown of
heterochromatin foci (SAHF) repressing pro-proliferative E2F tar-
the mitochondrial calcium uniporter MCU fosters escape from
gets, both participating in the implementation of cell cycle arrest
senescence [16]. The ER and mitochondria can be spatially and func-
(Fig. 1) [1,2,5,6].
tionally coupled through mitochondria-associated ER membranes
Calcium critically controls many molecular processes and cel-
(MAMs) which favor the transfer of calcium from the ER to mito-
lular functions and there is accumulating evidence indicating that
chondria [23]. A role for these structures in the regulation of cellular
calcium signaling plays a key role in cellular senescence. We will
senescence can be speculated but remains to be assessed.
present the current knowledge on its implication in senescence,
as well as in the main senescence-associated features and in some
physiopathological contexts in which senescence is involved.
2.3. SASP regulation by calcium

2. Direct evidence of the role of calcium in senescence The observed rise in intracellular calcium in response to
senescence-inducing signals regulates the SASP through inter-
2.1. Increase in intracellular calcium levels in senescent cells leukin 1␣ (IL1␣), a pro-inflammatory cytokine which acts as a key
SASP initiator [24]. IL1␣ is synthesized as a precursor and then pro-
An elevation of intracellular calcium levels has been observed cessed by the calcium-activated protease calpain. In senescent cells,
in response to different types of senescence-inducing stresses the rise in intracellular levels of calcium, which is the co-factor of
(telomere shortening, oncogene activation, rotenone or oxidative calpain, activates this protease. This leads to increased IL1␣ pro-
stress) and in several cell types. This increase in calcium con- teolytic cleavage and enhanced expression of downstream SASP
centration has been attributed to calcium influx though plasma cytokines, such as IL6 and IL8.

Descargado para Anonymous User (n/a) en University of Chile de ClinicalKey.es por Elsevier en febrero 04, 2021.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
18 N. Martin, D. Bernard / Cell Calcium 70 (2018) 16–23

TRIGGERS

Telomere Oncogene Oxidave DNA Proteotoxic Other


shortening acvaon stress damage stress stresses

MAIN EFFECTORS

p53 and RB NF B mTOR


pathway pathway pathway

ARF IKK TSC1 TSC2


ATM/ATR

p53

RHEB1
IB
p21 p16
RELA p50

CDKs

mTORC1
RB RELA p50

S6K 4EBP1
E2F

E2F target genes NF B target genes Protein synthesis

FEATURES

Cell morphology Cell cycle SA-secretory Reacve oxygen


changes arrest phenotype (SASP) species (ROS)

SA- -galactosidase Sustained DNA


acvity damage

PHYSIOPATHOLOGICAL EFFECTS

Fig. 1. Main molecular basis of cellular senescence. A wide variety of stresses activate effector pathways implementing cellular senescence. Note that only the main and best
characterized effectors are depicted here and that these pathways intricately interplay. Their activation leads to the establishment of senescence features, among which the
cell cycle arrest and the senescence-associated secretory phenotype (SASP) are the main features causing physiopathological effects.

2.4. Role of the calcium/NFAT signaling pathway in senescence increase, calmodulin, a calcium sensor protein, binds to calcium
and activates calcineurin. This phosphatase triggers NFAT dephos-
The calcium-dependent transcription factor NFAT (nuclear phorylation and its translocation to the nucleus, where it regulates
factor of activated T-cells) is activated through the cal- the expression of its target genes [26]. Several studies have shown
cium/calmodulin/calcineurin pathway [25]. Upon cytosolic calcium that NFAT plays a crucial role in cellular senescence. Indeed the

Descargado para Anonymous User (n/a) en University of Chile de ClinicalKey.es por Elsevier en febrero 04, 2021.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
N. Martin, D. Bernard / Cell Calcium 70 (2018) 16–23 19

Stress
Calcium
channels

Cytosol

Ca2+
IP3R
Endoplasmic
reculum
IP3R

MCU
Mitochondria
Calmodulin Calpain

Calcineurin

ROS NFAT IL1

Nucleus

Cell cycle arrest SASP

CELLULAR SENESCENCE

Fig. 2. Known roles of calcium in cellular senescence. In response to senescence-inducing stresses, intracellular calcium levels increase and promote senescence though
several calcium-dependent mechanisms triggering cell cycle arrest and SASP production. Only direct roles of calcium in senescence are depicted here.

calcineurin/NFAT signaling pathway is critically required for p53- 3. Other roles for calcium in the main
dependent senescence in skin [27]. NFATc1 directly represses the senescence-associated features
expression of ATF3, a transcription factor related to the AP-1
family which inhibits the expression of p53 and other senescence- 3.1. Cell proliferation
associated genes. Impairment of the calcineurin/NFAT pathway
suppresses p53-dependent senescence and promotes tumor for- One of the two functionally crucial features of senescent cells is
mation of H-RASV12 -expressing primary human keratinocytes or their state of stable proliferation arrest. Cell proliferation is directly
keratinocyte-derived squamous cell carcinoma cells. However, it linked to cell cycle progression. Calcium signaling plays a key role
has also been shown that NFATc1 activation in prostate epithelium throughout the mammalian cell cycle. Intracellular calcium con-
promotes prostatic intraepithelial neoplasia and prostate adeno- centration is modulated during the G1 phase, G1/S transition and
carcinoma [28]. Moreover, NFATc1 accelerates PTEN null-driven mitosis [31]. Calcium is important in early G1 for the expression
prostate tumorigenesis. NFATc1 overcomes PTEN loss-induced of genes such as FOS, JUN and MYC and later for RB phospho-
cellular senescence by downregulating the cell cycle inhibitor rylation. Antagonists of calmodulin trigger cell cycle arrest in G1
p21 [28]. Altogether, it seems that the calcineurin/NFAT path- [32]. Calmodulin kinase (CaMK) and calcineurin, downstream tar-
way displays senescence-promoting or –suppressing functions gets of calmodulin, are required for cyclin D1 expression [33,34].
depending on the cell type and context. Interestingly, NFATc1 The calcium/calmodulin pathway regulates several transcription
has also been described to activate the expression of the ER cal- factors such as NFAT or the cAMP responsive element binding
cium release channel IP3R2, which could be an additional way protein (CREB) which target genes that control cell cycle pro-
for the calcineurin/NFAT pathway to impact senescence [29]. Of gression from G1 to S. Direct upregulation of MYC expression by
course, calcium/calmodulin/calcineurin/NFAT pathway can also calcineurin-activated NFATc1 induces an increase in the expression
exert effects independently of its effect on cellular senescence, such of MYC target genes, cyclin E and E2F [35]. CREB undergoes calcium-
as promoting T cell clonal expansion [30]. induced phosphorylation by CAMK2 and 4. Once phosphorylated
In addition to these reports which clearly demonstrate a direct CREB binds to the cAMP response element (CRE) on the promoter of
role for calcium signaling in cellular senescence (Fig. 2), there is target genes, such as cyclin D1, and recruits co-activators, includ-
extensive literature on the involvement of calcium in features asso- ing the histone acetyl-transferases CBP and p300 which activate
ciated with senescence. The main implications of calcium in this gene transcription [36]. CDK2 activity is also increased by cal-
context will be reviewed in the next section. cineurin. In addition, the calcium/calmodulin pathway promotes
RB phosphorylation by enhancing p34 Cdc2 kinase (CDK1) activa-

Descargado para Anonymous User (n/a) en University of Chile de ClinicalKey.es por Elsevier en febrero 04, 2021.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
20 N. Martin, D. Bernard / Cell Calcium 70 (2018) 16–23

tion [37]. P53, similarly to RB, may also be regulated by calcium.


P53 was shown to bind to several members of the S100 family of INTRACELLULAR
calcium-binding proteins in a calcium-dependent manner. These CALCIUM
interactions can regulate the stability and transcriptional activity
of p53. For instance, S100B binds to p53 in melanoma, reduces
p53 protein levels and thus represses the function of p53 [38]. The
TIMELY REGULATED INCREASED
interaction between p53 and some calcium-binding proteins may concentraon concentraon
thus influence cell cycle progression. However, a comprehensive
analysis of the direct impact of calcium on the two key master regu-
lators of cellular senescence p53 and RB in primary cells is currently
lacking. During mitosis, a rise in cytosolic calcium at the onset of Cells with impaired
anaphase, just preceding nuclear envelope breakdown, has been senescence and death Normal cells
observed in cultured mammalian fibroblasts and epithelial cells effector programs
as well as in embryos [39–41]. Mimicking this cytosolic calcium
increase by artificially injecting calcium into the embryo causes
early nuclear envelope breakdown, whereas chelating it prevents
this breakdown and suspends mitosis [40]. In addition, defects PRO-PROLIFERATIVE and CYTOSTATIC and
in calcium- and CAMK2-mediated control of centrosome duplica- PRO-SURVIVAL effects CYTOTOXIC effects
tion (in G1/S) and separation (in G2/M) result in aberrant mitotic Cell cycle progression
Cellular senescence
spindles, genetic instability and aneuploidy [42,43]. Modulation of
Migraon/invasion
cytosolic calcium concentration is therefore a crucial regulatory
Metabolism Cell death
step for cell cycle progression. Calcium signaling is not only piv-
otal for cell cycle progression but more generally for the survival
and proper functioning of cells. For example, calcium is required
Fig. 3. Dual effects of calcium on cellular life. Calcium has pleiotropic effects on
for energy production and acts at several levels in the mitochon- cellular life depending on its intracellular levels, the regulation of these levels and
dria to stimulate ATP synthesis. In addition, calcium controls many the status of the cells.
other aspects of cellular metabolism [44]. In conclusion, calcium is
required for cell cycle progression, proliferation and survival.
to promote the secretion of IL1␤, another component of the SASP.
These latter observations appear to be contradictory with
Calmodulin binds to IL1␤ precursors and IL1␤-processing and
the pro-senescence effect of calcium described in Section 2.
–release depends on this calcium-mediated interaction [54].
We can speculate that a spatio-temporal elevation of calcium
For the sake of clarity we have only discussed the role of calcium
concentration might switch the effect of calcium from a pro-
signaling on cell proliferation and SASP, the two main hallmarks
proliferative/survival effect to a cyto-static/toxic effect. Beyond
of senescent cells explaining its role in various physiopatholog-
the literature presented in Section 2, showing an involvement of
ical responses. However, calcium signaling plays a key role in
calcium in senescence, a rapid rise in mitochondrial calcium lev-
many other senescence-associated features such as DNA damage
els precedes cell death and contributes to its induction [45,46].
response and autophagy, suggesting that calcium may also impact
Changes in calcium homeostasis may therefore drive cells to enter
senescence in many additional ways than the ones we focused on.
either senescence or cell death, probably depending on the charac-
teristics of the spatio-temporal elevation of calcium concentrations
(site and level of accumulation as well as kinetic and duration of the 4. Role of calcium in senescence-associated
increase). physiopathological conditions
Still in some circumstances increased calcium might not exert
cytostatic/toxic effects if the effector pathways like p53, RB or 4.1. Role of cellular senescence in vivo
caspases are not functional. It is tempting to speculate that it is
what happens in cancer cells, where these pathways are altered Senescent cells accumulate with age and contribute to age-
and increased calcium reported. Then in these specific conditions related diseases and organismal aging. Although a matter of debate
increased calcium might promote survival, proliferation, migration for many years, this role for senescence has definitely been demon-
and invasion phenotypes (Fig. 3) [47–49]. strated the last few years by the evidence that elimination of
senescent cells during accelerated or normal aging extends life
3.2. Senescence-associated secretory phenotype span and improves numerous age-associated pathologies such as
atherosclerosis, type II diabetes, glomerulosclerosis, cardiac aging,
Beside the cell proliferation arrest, the other functionally crucial lung degenerative diseases, cancer relapse and progression, as well
feature of senescent cells is their distinct pro-inflammatory secre- as immunosenescence [8,55–61].
tome (SASP). The SASP is regulated at the transcriptional level by Senescent cells are also observed in the organism at various
the NF␬B pathway, which induces the expression of SASP compo- stages of life during which senescence can have beneficial health
nents such as IL6 and IL8 pro-inflammatory cytokines. Activation effects. Cellular senescence is activated during normal embry-
of the NF␬B pathway depends on intracellular free calcium under onic development, where it contributes to embryonic growth and
certain circumstances. Activation of calmodulin has been shown patterning [62,63]. It is also involved in various responses, from
to inhibit c-REL and allow RELA to translocate into the nucleus promoting wound healing [64] to increasing insulin secretion [65].
[50–52]. In the context of tumor initiation, cellular senescence is a major
The SASP is also induced at the translational level by the mTOR failsafe program that is activated upon oncogenic stresses, short
pathway. It has recently been demonstrated that mTORC1 acti- telomeres and mutagenic signals, to eliminate cells at risk of
vation requires lysosomal calcium release through the calcium transformation. In benign preneoplastic lesions, oncogene-induced
channel TRPML1. Lysosomal calcium activates mTORC1 through senescence (OIS) limits the proliferation of these cells and triggers
calmodulin, which associates with mTORC1 and stimulates its their clearance by the immune system. OIS is therefore a potent
kinase activity [53]. Calcium and calmodulin have also been shown protective barrier in the initial steps of tumorigenesis [9,66,67].

Descargado para Anonymous User (n/a) en University of Chile de ClinicalKey.es por Elsevier en febrero 04, 2021.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
N. Martin, D. Bernard / Cell Calcium 70 (2018) 16–23 21

2 kinase (phospho-Chk2) are increased in this pathology [74]. An


upregulation of intracellular calcium concentration and a calcium-
CELLULAR
dependent activation of the NF␬B pathway, which is a key SASP
SENESCENCE
regulator, are also observed in cystic fibrosis airway epithelial cells
[75]. These findings suggest that senescence could be implicated
in this disease and that calcium could regulate senescence in this
TIMELY REGULATED CHRONIC context.
senescence senescence It has been shown that p16-induced senescence in pancre-
atic ␤ cells enhances insulin secretion, partly through mTOR and
the peroxisome proliferator-activated receptor ␥ (PPAR␥) [65].
␤ cell-specific deletion of the calcineurin regulatory subunit cal-
cineurin b1 provokes age-dependent diabetes in mice due to
BENEFICIAL DETRIMENTAL decreased ␤ cell proliferation, decreased pancreatic insulin content
effects effects and hypoinsulinaemia. Consistently, conditional NFAT activation in
normal adult ␤ cells increases their proliferation and mass, provok-
Embryonic development Cancer relapse and progression ing hyperinsulinemia, and induces the expression of genes which
are critical for the endocrine function of ␤ cells [76]. Thus, calcium
Type II diabetes
Atherosclerosis
signaling and ␤-cell senescence promotes insulin secretion.
Wound healing Even though senescence appears to promote ␤-cell insulin
Glomerulosclerosis
Cardiac aging secretion, senescence in insulin sensitive organs might render
Lung degenerave diseases
Tumor suppression Immunosenescence
them insulin resistant. Indeed, high fat diets and obesity, which
are known to result in insulin resistance, promote senescence of
adipocytes and hepatocytes, two cell types responding to insulin to
metabolize circulating glucose. Decreasing senescence by deplet-
Fig. 4. Dual effects of cellular senescence on organismal health. Based on its dual ing p53 restores the response to insulin, demonstrating that in
effects, cellular senescence has been proposed to be an example of antagonistic that given context senescence provokes insulin resistance [60,77].
pleiotropy. Accumulation of mitochondrial calcium is observed during obesity
and has been shown to promote insulin resistance [78], as does
Altogether age-related chronic senescence is thought to exert accumulation of senescent cells, once again supporting a functional
detrimental effects on health, whereas timely-regulated senes- link between calcium, and in particular mitochondrial calcium, and
cence is thought to exert beneficial effects (Fig. 4) [57,68,69]. senescence. However, the role of calcium is not that simple as
cytosolic calcium can both decrease or increase insulin sensitiv-
4.2. Implication of calcium in some physiopathological responses ity [79,80]. We can speculate that the effect of calcium on insulin
controlled by senescence sensitivity depends on its spatio-temporal regulation and then on
its ability to induce or not senescence as previously discussed.
Calcium plays a pivotal role in several physiopathological con- Together, calcium might exert some of its effects on health and
texts in which senescence is involved, suggesting a functional diseases through its ability to control cellular senescence.
link between calcium and senescence in vivo. Cellular senescence
is critically implicated in wound healing. Appearance of senes-
cent fibroblasts and endothelial cells is observed soon after skin 5. Concluding remarks
wounding. These cells accelerate wound closure by triggering
myofibroblast differentiation through the secretion of platelet- Cellular senescence is a cellular response to a wide variety of
derived growth factor AA (PDGF-AA) [64]. Calcium is an essential stresses that plays a key role in development, aging and diseases.
regulator of wound responses [70]. Epithelial wounds trigger inter- Identifying the molecular and cellular mechanisms controlling this
cellular calcium waves which propagate through the wounded process is currently a key challenge to improve our understanding
epithelium. Wounds induce an elevation of cytoplasmic calcium of these physiopathological conditions and be able to envision ways
levels through the opening of plasma membrane channels such to impact them. In the last decade the concept that calcium signal-
as TRPs or through calcium release from intracellular stores. ing is a key regulator of cellular senescence has emerged. Thus,
This increase in intracellular calcium concentrations induces ROS calcium signaling appears to be an important effector of cellular
production by activating the epithelial NADPH-oxidase DUOX senescence.
or by triggering mitochondrial permeability transition. Calcium However, it is very likely that our current knowledge is only the
also guides the secretion of paracrine mediators and is required tip of the iceberg and that many more implications of calcium sig-
for epithelial sheet motility and integrity. In addition to their naling in cellular senescence will be discovered in the near future.
coordinated role during wound healing, migration and invasion To date only a few members of the calcium signaling pathway have
contribute to tumor progression. Senescent cells owing to their been identified as senescence regulators. Calcium signaling impli-
SASP have been proposed to favor cell migration and invasion of cates many players and signaling cascades. According to the KEGG
neighboring tissues [8]. In particular, senescent fibroblasts in the pathway database, 180 proteins participate in the calcium signal-
tumor stroma promote, through their SASP, cancer cell migration ing pathway. It includes a large variety of proteins, among which
[71–73]. Calcium directly controls the cell migration process, by numerous calcium channels from diverse families and calcium-
impacting cytoskeleton organization as well as signaling pathways binding proteins. Assessing the involvement of each of these factors
[47]. Altogether, this suggests that calcium can promote cell migra- in cellular senescence would improve our understanding of the
tion and contribute to the pro-migratory effect of the SASP, thus role of calcium signaling in this process. To this end, regulation of
impacting neighboring cells. their expression and activity as well as their functional impact on
Cystic fibrosis is a genetic lung disease characterized by senescence should be investigated.
chronic airway inflammation. Levels of senescence markers such With regards to a more applied perspective, dissecting the role
as p16, phospho-histone H2A.X (␥H2A.X) and phospho-checkpoint of players of the calcium signaling pathway in senescence may

Descargado para Anonymous User (n/a) en University of Chile de ClinicalKey.es por Elsevier en febrero 04, 2021.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
22 N. Martin, D. Bernard / Cell Calcium 70 (2018) 16–23

reveal targets of interest for medical purposes. This could allow [26] P.G. Hogan, L. Chen, J. Nardone, A. Rao, Transcriptional regulation by calcium,
the identification of new early diagnostic markers and shed light calcineurin, and NFAT, Genes Dev. 17 (2003) 2205–2232.
[27] X. Wu, B.C. Nguyen, P. Dziunycz, et al., Opposing roles for calcineurin and
on innovative therapeutic strategies. Indeed, impacting senescence ATF3 in squamous skin cancer, Nature 465 (2010) 368–372.
can be beneficial in some physiopathological contexts such as [28] K.R. Manda, P. Tripathi, A.C. Hsi, et al., NFATc1 promotes prostate
aging and aging-related diseases. A possible avenue for modulating tumorigenesis and overcomes PTEN loss-induced senescence, Oncogene 35
(2016) 3282–3292.
senescence could be to manipulate calcium levels or calcium sig- [29] N. Sankar, P.P. deTombe, G.A. Mignery, Calcineurin-NFATc regulates type 2
naling. In this context calcium channels would be good druggable inositol 1,4,5-trisphosphate receptor (InsP3R2) expression during cardiac
candidates to target. remodeling, J. Biol. Chem. 289 (2014) 6188–6198.
[30] Y. Gwack, S. Feske, S. Srikanth, P.G. Hogan, A. Rao, Signalling to transcription:
store-operated Ca2+ entry and NFAT activation in lymphocytes, Cell Calcium
42 (2007) 145–156.
Acknowledgement [31] G. Pande, N.A. Kumar, P.S. Manogaran, Flow cytometric study of changes in the
intracellular free calcium during the cell cycle, Cytometry 24 (1996) 55–63.
This work has been supported by the Fondation ARC pour la [32] C.D. Rasmussen, A.R. Means, Calmodulin is required for cell-cycle progression
during G1 and mitosis, EMBO J. 8 (1989) 73–82.
recherche sur le cancer.
[33] C.R. Kahl, A.R. Means, Calcineurin regulates cyclin D1 accumulation in
growth-stimulated fibroblasts, Mol. Biol. Cell 15 (2004) 1833–1842.
[34] T.A. Morris, R.J. DeLorenzo, R.M. Tombes, CaMK-II inhibition reduces cyclin D1
References levels and enhances the association of p27kip1 with Cdk2 to cause G1 arrest
in NIH 3T3 cells, Exp. Cell Res. 240 (1998) 218–227.
[1] T. Kuilman, C. Michaloglou, W.J. Mooi, D.S. Peeper, The essence of senescence, [35] M. Buchholz, A. Schatz, M. Wagner, et al., Overexpression of c-myc in
Genes Dev. 24 (2010) 2463–2479. pancreatic cancer caused by ectopic activation of NFATc1 and the
[2] F. Rodier, J. Campisi, Four faces of cellular senescence, J. Cell Biol. 192 (2011) Ca2+/calcineurin signaling pathway, EMBO J. 25 (2006) 3714–3724.
547–556. [36] G. Schneider, F. Oswald, C. Wahl, F.R. Greten, G. Adler, R.M. Schmid,
[3] L. Hayflick, P.S. Moorhead, The serial cultivation of human diploid cell strains, Cyclosporine inhibits growth through the activating transcription
Exp. Cell Res. 25 (1961) 585–621. factor/cAMP-responsive element-binding protein binding site in the cyclin D1
[4] J. Campisi, L. Robert, Cell senescence: role in aging and age-related diseases, promoter, J. Biol. Chem. 277 (2002) 43599–43607.
Interdiscip. Top. Gerontol. 39 (2014) 45–61. [37] N. Takuwa, W. Zhou, M. Kumada, Y. Takuwa, Ca(2+)-dependent stimulation of
[5] I. Ben Porath, R.A. Weinberg, The signals and pathways activating cellular retinoblastoma gene product phosphorylation and p34cdc2 kinase activation
senescence, Int. J. Biochem. Cell Biol. 37 (2005) 961–976. in serum-stimulated human fibroblasts, J. Biol. Chem. 268 (1993) 138–145.
[6] R. Salama, M. Sadaie, M. Hoare, M. Narita, Cellular senescence and its effector [38] J. Lin, Q. Yang, P.T. Wilder, F. Carrier, D.J. Weber, The calcium-binding protein
programs, Genes Dev. 28 (2014) 99–114. S100 B down-regulates p53 and apoptosis in malignant melanoma, J. Biol.
[7] J.C. Acosta, A. O’Loghlen, A. Banito, et al., Chemokine signaling via the CXCR2 Chem. 285 (2010) 27487–27498.
receptor reinforces senescence, Cell 133 (2008) 1006–1018. [39] M. Poenie, J. Alderton, R. Steinhardt, R. Tsien, Calcium rises abruptly and
[8] J.P. Coppe, C.K. Patil, F. Rodier, et al., Senescence-associated secretory briefly throughout the cell at the onset of anaphase, Science 233 (1986)
phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the 886–889.
p53 tumor suppressor, PLoS Biol. 6 (2008) 2853–2868. [40] R.A. Steinhardt, J. Alderton, Intracellular free calcium rise triggers nuclear
[9] T.W. Kang, T. Yevsa, N. Woller, et al., Senescence surveillance of pre-malignant envelope breakdown in the sea urchin embryo, Nature 332 (1988) 364–366.
hepatocytes limits liver cancer development, Nature 479 (2011) 547–551. [41] R.M. Tombes, G.G. Borisy, Intracellular free calcium and mitosis in
[10] J.C. Acosta, A. Banito, T. Wuestefeld, et al., A complex secretory program mammalian cells: anaphase onset is calcium modulated, but is not triggered
orchestrated by the inflammasome controls paracrine senescence, Nat. Cell by a brief transient, J. Cell Biol. 109 (1989) 627–636.
Biol. 15 (2013) 978–990. [42] Y. Matsumoto, J.L. Maller, Calcium, calmodulin, and CaMKII requirement for
[11] N. Herranz, S. Gallage, M. Mellone, et al., mTOR regulates MAPKAPK2 initiation of centrosome duplication in Xenopus egg extracts, Science 295
translation to control the senescence-associated secretory phenotype, Nat. (2002) 499–502.
Cell Biol. 17 (2015) 1205–1217. [43] W.Y. Tsang, A. Spektor, D.J. Luciano, et al., CP110 cooperates with two
[12] T. Kuilman, C. Michaloglou, L.C.W. Vredeveld, et al., Oncogene-induced calcium-binding proteins to regulate cytokinesis and genome stability, Mol.
senescence relayed by an interleukin-dependent inflammatory network, Cell Biol. Cell 17 (2006) 3423–3434.
133 (2008) 1019–1031. [44] B. Glancy, R.S. Balaban, Role of mitochondrial Ca2+ in the regulation of
[13] R.M. Laberge, Y. Sun, A.V. Orjalo, et al., MTOR regulates the pro-tumorigenic cellular energetics, Biochemistry 51 (2012) 2959–2973.
senescence-associated secretory phenotype by promoting IL1A translation, [45] G. Hajnoczky, E. Davies, M. Madesh, Calcium signaling and apoptosis,
Nat. Cell Biol. 17 (2015) 1049–1061. Biochem. Biophys. Res. Commun. 304 (2003) 445–454.
[14] M. Ferrand, O. Kirsh, A. Griveau, et al., Screening of a kinase library reveals [46] S.S. Smaili, Y.T. Hsu, R.J. Youle, J.T. Russell, Mitochondria in Ca2+ signaling and
novel pro-senescence kinases and their common NF-kappaB-dependent apoptosis, J. Bioenerg. Biomembr. 32 (2000) 35–46.
transcriptional program, Aging (Albany N.Y.) 7 (2015) 986–1003. [47] N. Prevarskaya, R. Skryma, Y. Shuba, Calcium in tumour metastasis: new roles
[15] X. Yu, X. Li, G. Jiang, et al., Isradipine prevents rotenone-induced intracellular for known actors, Nat. Rev. Cancer 11 (2011) 609–618.
calcium rise that accelerates senescence in human neuroblastoma SH-SY5Y [48] N. Prevarskaya, R. Skryma, Y. Shuba, Ion channels and the hallmarks of cancer,
cells, Neuroscience 246 (2013) 243–253. Trends Mol. Med. 16 (2010) 107–121.
[16] C. Wiel, H. Lallet-Daher, D. Gitenay, et al., Endoplasmic reticulum calcium [49] N. Prevarskaya, H. Ouadid-Ahidouch, R. Skryma, Y. Shuba, Remodelling of
release through ITPR2 channels leads to mitochondrial calcium accumulation Ca2+ transport in cancer: how it contributes to cancer hallmarks? Philos.
and senescence, Nat. Commun. 5 (2014) 3792. Trans. R Soc. Lond. B Biol. Sci. 369 (2014) 20130097.
[17] A.V. Borodkina, A.N. Shatrova, P.I. Deryabin, et al., Calcium alterations signal [50] J.E. Kim, S.Y. Kim, S.Y. Lim, E. Kieff, Y.J. Song, Role of
either to senescence or to autophagy induction in stem cells upon oxidative Ca2+/calmodulin-dependent kinase II-IRAK1 interaction in LMP1-induced
stress, Aging (Albany N.Y.) 8 (2016) 3400–3418. NF-kappaB activation, Mol. Cell. Biol. 34 (2014) 325–334.
[18] H. Akl, G. Bultynck, Altered Ca(2+) signaling in cancer cells: proto-oncogenes [51] H. Ling, C.B. Gray, A.C. Zambon, et al., Ca2+/Calmodulin-dependent protein
and tumor suppressors targeting IP3 receptors, Biochim. Biophys. Acta 1835 kinase II delta mediates myocardial ischemia/reperfusion injury through
(2013) 180–193. nuclear factor-kappaB, Circ. Res. 112 (2013) 935–944.
[19] M.T. Khan, L. Wagner, D.I. Yule, C. Bhanumathy, S.K. Joseph, Akt kinase [52] G. Maubach, O. Sokolova, M. Wolfien, H.J. Rothkotter, M. Naumann,
phosphorylation of inositol 1,4,5-trisphosphate receptors, J. Biol. Chem. 281 Ca2+/calmodulin-dependent kinase II contributes to inhibitor of nuclear
(2006) 3731–3737. factor-kappa B kinase complex activation in Helicobacter pylori infection, Int.
[20] T. Szado, V. Vanderheyden, J.B. Parys, et al., Phosphorylation of inositol J. Cancer 133 (2013) 1507–1512.
1,4,5-trisphosphate receptors by protein kinase B/Akt inhibits Ca2+ release [53] R.J. Li, J. Xu, C. Fu, et al., Regulation of mTORC1 by lysosomal calcium and
and apoptosis, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 2427–2432. calmodulin, Elife 5 (2016).
[21] G. Ferbeyre, E. de Stanchina, E. Querido, N. Baptiste, C. Prives, S.W. Lowe, PML [54] J.S. Ainscough, G.F. Gerberick, I. Kimber, R.J. Dearman, Interleukin-1beta
is induced by oncogenic ras and promotes premature senescence, Genes Dev. processing is dependent on a calcium-mediated interaction with calmodulin,
14 (2000) 2015–2027. J. Biol. Chem. 290 (2015) 31151–31161.
[22] C. Giorgi, K. Ito, H.K. Lin, et al., PML regulates apoptosis at endoplasmic [55] D.J. Baker, B.G. Childs, M. Durik, et al., Naturally occurring p16(Ink4a)-positive
reticulum by modulating calcium release, Science 330 (2010) 1247–1251. cells shorten healthy lifespan, Nature 530 (2016) 184–189.
[23] T. Hayashi, R. Rizzuto, G. Hajnoczky, T.P. Su, MAM: more than just a [56] J. Chang, Y. Wang, L. Shao, et al., Clearance of senescent cells by ABT263
housekeeper, Trends Cell Biol. 19 (2009) 81–88. rejuvenates aged hematopoietic stem cells in mice, Nat. Med. 22 (2016)
[24] D.A. McCarthy, R.R. Clark, T.R. Bartling, M. Trebak, J.A. Melendez, Redox 78–83.
control of the senescence regulator interleukin-1alpha and the secretory [57] B.G. Childs, D.J. Baker, J.L. Kirkland, Campisi J, van Deursen JM. Senescence and
phenotype, J. Biol. Chem. 288 (2013) 32149–32159. apoptosis: dueling or complementary cell fates, EMBO Rep. 15 (2014)
[25] G.R. Crabtree, E.N. Olson, NFAT signaling: choreographing the social lives of 1139–1153.
cells, Cell 109 (Suppl) (2002) S67–S79.

Descargado para Anonymous User (n/a) en University of Chile de ClinicalKey.es por Elsevier en febrero 04, 2021.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
N. Martin, D. Bernard / Cell Calcium 70 (2018) 16–23 23

[58] M. Demaria, M.N. O’Leary, J. Chang, et al., Cellular senescence promotes [71] I. Aifuwa, A. Giri, N. Longe, S.H. Lee, S.S. An, D. Wirtz, Senescent stromal cells
adverse effects of chemotherapy and cancer relapse, Cancer Discov. 7 (2017) induce cancer cell migration via inhibition of RhoA/ROCK/myosin-based cell
165–176. contractility, Oncotarget 6 (2015) 30516–30531.
[59] M. Hashimoto, A. Asai, H. Kawagishi, et al., Elimination of p19ARF-expressing [72] V. Farsam, A. Basu, M. Gatzka, et al., Senescent fibroblast-derived Chemerin
cells enhances pulmonary function in mice, JCI Insight 1 (2016) e87732. promotes squamous cell carcinoma migration, Oncotarget 7 (2016)
[60] T. Minamino, M. Orimo, I. Shimizu, et al., A crucial role for adipose tissue p53 83554–83569.
in the regulation of insulin resistance, Nat. Med. 15 (2009) 1082–1087. [73] P.A. Toste, A.H. Nguyen, B.E. Kadera, et al., Chemotherapy-induced
[61] M.J. Schafer, T.A. White, K. Iijima, et al., Cellular senescence mediates fibrotic inflammatory gene signature and protumorigenic phenotype in pancreatic
pulmonary disease, Nat. Commun. 8 (2017) 14532. CAFs via stress-associated MAPK, Mol. Cancer Res. 14 (2016) 437–447.
[62] D. Munoz-Espin, M. Canamero, A. Maraver, et al., Programmed cell senescence [74] B.M. Fischer, J.K. Wong, S. Degan, et al., Increased expression of senescence
during mammalian embryonic development, Cell 155 (2013) 1104–1118. markers in cystic fibrosis airways, Am. J. Physiol. Lung Cell. Mol. Physiol. 304
[63] M. Storer, A. Mas, A. Robert-Moreno, et al., Senescence is a developmental (2013) L394–L400.
mechanism that contributes to embryonic growth and patterning, Cell 155 [75] O. Tabary, E. Boncoeur, R. de Martin, et al., Calcium-dependent regulation of
(2013) 1119–1130. NF-(kappa)B activation in cystic fibrosis airway epithelial cells, Cell. Signal. 18
[64] M. Demaria, N. Ohtani, S.A. Youssef, et al., An essential role for senescent cells (2006) 652–660.
in optimal wound healing through secretion of PDGF-AA, Dev. Cell 31 (2014) [76] J.J. Heit, A.A. Apelqvist, X. Gu, et al., Calcineurin/NFAT signalling regulates
722–733. pancreatic beta-cell growth and function, Nature 443 (2006) 345–349.
[65] A. Helman, A. Klochendler, N. Azazmeh, et al., p16(Ink4a)-induced senescence [77] X. Zhang, D. Zhou, R. Strakovsky, Y. Zhang, Y.X. Pan, Hepatic cellular
of pancreatic beta cells enhances insulin secretion, Nat. Med. 22 (2016) senescence pathway genes are induced through histone modifications in a
412–420. diet-induced obese rat model, Am. J. Physiol. Gastrointest. Liver Physiol. 302
[66] M. Collado, M. Serrano, The senescent side of tumor suppression, ABBV Cell (2012) G558–G564.
Cycle 4 (2005). [78] A.P. Arruda, B.M. Pers, G. Parlakgul, E. Guney, K. Inouye, G.S. Hotamisligil,
[67] M. Collado, M. Serrano, Senescence in tumours: evidence from mice and Chronic enrichment of hepatic endoplasmic reticulum-mitochondria contact
humans, Nat. Rev. Cancer 10 (2010) 51–57. leads to mitochondrial dysfunction in obesity, Nat. Med. 20 (2014)
[68] D. Munoz-Espin, M. Serrano, Cellular senescence: from physiology to 1427–1435.
pathology, Nat. Rev. Mol. Cell Biol. 15 (2014) 482–496. [79] J.T. Lanner, A. Katz, P. Tavi, et al., The role of Ca2+ influx for insulin-mediated
[69] P.A. Perez-Mancera, A.R. Young, M. Narita, Inside and out: the activities of glucose uptake in skeletal muscle, Diabetes 55 (2006) 2077–2083.
senescence in cancer, Nat. Rev. Cancer 14 (2014) 547–558. [80] D.S. Worrall, J.M. Olefsky, The effects of intracellular calcium depletion on
[70] B. Enyedi, P. Niethammer, Mechanisms of epithelial wound detection, Trends insulin signaling in 3T3-L1 adipocytes, Mol. Endocrinol. 16 (2002) 378–389.
Cell Biol. 25 (2015) 398–407.

Descargado para Anonymous User (n/a) en University of Chile de ClinicalKey.es por Elsevier en febrero 04, 2021.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.

You might also like