Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

REVIEW

Connective Tissue and Fibroblast


Senescence in Skin Aging
Meinhard Wlaschek1, Pallab Maity1, Evgenia Makrantonaki1,2 and Karin Scharffetter-Kochanek1
There is increasing evidence that skin aging is signif- furthermore intends to provide insight into state-of-the-art
icantly enforced by the accumulation of senescent technology, which has undoubtedly advanced our under-
dermal fibroblasts. Various stressors damaging mac- standing of fibroblast senescence driving skin and other organ
romolecules inside and outside fibroblasts are aging with the unique opportunity to develop and refine
responsible. In addition, NK cells fail to adequately modern preventive and therapeutic strategies.
remove senescent (SEN) fibroblasts from tissues. SEN
fibroblasts by the release of the proinflammatory, Senescence of fibroblasts in skin aging
tissue degrading senescent-associated secretory
The role of senescence in skin aging. An emerging
phenotype factors and vesicles with distinct cargo
hypothesis of skin aging postulates that mainly fibroblast
impact on their endogenous niche and spread senescence drives skin decline and skin aging due to irre-
senescence and skin aging. In this review, we will versible proliferation arrest and enhanced release of a
further discuss less noticed facets, including the senescence-associated secretory phenotype (SASP). SASP,
plasticity of distinct dermal fibroblast phenotypes, the through chemokines and proinflammatory factors, induces
underestimated impact of the extracellular matrix it- chronic inflammation, reduces proliferation by impaired
self, and the depletion of fibroblast subsets on skin release of essential GFs, and enhances the degradation of the
homeostasis and aging. ECM (connective tissue) by enhanced activation of proteo-
Journal of Investigative Dermatology (2021) 141, 985e992; doi:10.1016/ lytic enzymes, including matrix-degrading metal-
j.jid.2020.11.010 loproteinases. Cellular senescence transiently occurs in
several physiological conditions during embryogenesis
(Muñoz-Espı́n and Serrano, 2014) and wound healing
(Demaria et al., 2014) shaping and regenerating organs. By
Introduction contrast, fibroblast senescence is mainly a persisting perma-
Dermal connective tissue connects all histogenetically nent state and, in consequence, drives skin aging, aging in
distinct cells and tissues of the skin to a functional organ. general and aging-related diseases (Campisi et al., 2019,
Fibroblasts constitute the principal component of the con- 2005; Demaria et al., 2015; Toutfaire et al., 2017; Treiber
nective tissue. As such, fibroblasts occur—apart from the et al., 2011; Velarde and Demaria, 2016; Wang and
skin—in virtually every tissue and organ of the body. The Dreesen, 2018). Given that the fibroblasts confined to the
capacity of fibroblasts to synthesize and organize the extra- connective tissue stabilize every organ parenchyma, cellular
cellular matrix (ECM) and communicate with the adjacent senescence of fibroblasts may play a more general role in
stem cell (SC) niche and quiescent, mitotic, and postmitotic organ aging, in organismal aging, and likely in many aging-
cells and tissues of distinct origin makes them, so far, an related diseases (Figure 1). In fact, senescent (SEN) fibro-
underestimated central component in skin homeostasis and blasts are increased in human skin, in the dermis (Dimri
aging. In this review, we will summarize the advanced un- et al., 1995; Ressler et al., 2006), and in many other organs
derstanding of the role of cellular senescence for stromal (Krishnamurthy et al., 2004; Tuttle et al., 2020). The most
aging in general as well as for skin aging in particular. important evidence for the role of SEN fibroblasts in enforc-
Wherever possible, we will differentiate between data that ing skin aging is summarized in Table 1. Permanent senes-
are relevant for aging in general and stromal aging and—most cence can be induced in quiescent (nonreplicating)
importantly—the ones that have truly been shown to play a fibroblasts in the skin by cell-autonomous and exogenous
role in murine and human skin aging. This review stressors leading to the shortening of the chromosomal
termini, termed telomeres (Bodnar et al., 1998; Harley et al.,
1
1990; Yu et al., 1990), mitochondrial dysfunction (Berneburg
Department of Dermatology and Allergic Diseases, Ulm University, Ulm,
Germany; and 2Dermatology Center Wildeshausen, Wildeshausen,
et al., 1997; Birch et al., 2018; Birch-Machin et al., 1998;
Germany Passos et al., 2007), DNA damage, and subsequent activation
Correspondence: Karin Scharffetter-Kochanek, Department of Dermatology of DNA damage response (DDR) signaling pathways leading
and Allergic Diseases, Ulm University, Albert-Einstein-Allee 23, 89081 Ulm, to permanent or transient cell cycle arrest (d’Adda di
Germany. E-mail: karin.scharffetter-kochanek@uniklinik-ulm.de Fagagna, 2008; Hoeijmakers, 2009) and metabolic reprog-
Abbreviations: AP-1, activator protein-1; CDK, cell cycle kinase; DDR, DNA ramming (da Silva and Schumacher, 2019; Figure 2). In this
damage response; ECM, extracellular matrix; MMP, matrix metalloproteinase;
MSC, mesenchymal stem cell; scRNA-seq, single cell RNA sequencing; SASP,
regard, also the accumulation of oxidized proteins due to the
senescence-associated secretory phenotype; SEN, senescent loss of the proteasomal capacity to remove oxidized and
Received 27 August 2020; revised 28 October 2020; accepted 11 November misfolded protein activates DDR pathways and thus leads to
2020; corrected proof published online 7 February 2021 the induction of cellular senescence (Chondrogianni and

ª 2021 The Authors. Published by Elsevier, Inc. on behalf of the Society for Investigative Dermatology. This is an open access
article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). www.jidonline.org 985
M Wlaschek et al.
Fibroblast Senescence and Skin Aging

Figure 1. SEN fibroblasts drive tissue,


organ, and organismal aging. SEN
fibroblasts imprint their environment
with a proinflammatory SASP, soluble
factors released upon the installment
of the senescence program, thereby
affecting distinct tissue in the skin and
various organs, here apart from the
skin, bone, and muscle among others.
This drives organ aging, aging-related
diseases, and likely organismal aging.
This hypothesis needs to be further
proven because it bears many
promising therapeutic and preventive
opportunities. This review gathers
further evidence. SASP, senescence-
associated secretory phenotype; SEN,
senescent.

Gonos, 2010) and to the release of soluble senescence- p15, or p27, which induce and maintain the state of senes-
spreading factors (Catalgol and Grune, 2009; Eckhart et al., cence, clearly depends on the specific triggers of senescence,
2019). Apart from many other stressors (summarized in including ROS imbalance (for review, see Lozano-Torres
Campisi [2005]; Gorgoulis et al. [2019]), mitochondrial et al. [2019]). In case of their inhibition, CDKs cannot
dysfunction in fibroblasts with increasing concentrations of phosphorylate Rb, which physiologically sequesters the
ROS (Gu et al., 2020; Rinnerthaler et al., 2015; Treiber et al., transcription factor E2F. E2F is a crucial protein enforcing the
2011) either leads to DNA damage of fibroblasts in the skin expression of genes that are mandatory for cell cycle pro-
and other organs (for review see Demaria et al. [2015]; gression. Its inhibition leads to cellular senescence.
Treiber et al. [2011]) or enhances activator protein-1 (AP-1)‒
and NF-kB‒dependent signaling pathways among others SASP and senescence spreading in skin aging. Clearly by
related to inflammation (Meyer et al., 2017). In fact, mito- their nonproliferating state with increased proteolytic activity
chondrial dysfunction is a major driver in both intrinsic and and suppressed function to deposit components of the ECM,
extrinsic skin aging and possibly constitutes the common link long-term SEN fibroblasts enforce tissue decline and skin
between both of them (Krutmann and Schroeder, 2009). aging. In addition, SEN fibroblasts have the unique property
To further explore the link between ROS and fibroblast not to die. They hardly can undergo apoptosis, cannot be
senescence, we earlier generated a mouse model with a sufficiently removed by the adaptive immune system, and, in
fibroblast specific deficiency for SOD2. Hence, mitochon- consequence, hang around in the connective tissue for a long
drial superoxide anions cannot be detoxified and, in conse- time with enhanced release of a variety of proinflammatory,
quence, accumulate in mitochondria of fibroblasts. These matrix-degrading chemokines, cytokines, and proteases and
fibroblast-specific, SOD2-deficient mice develop a severely a concomitantly suppressed release of essential GFs, collec-
accelerated skin aging phenotype with a marked reduction in tively referred to as SASP (Ghosh and Capell, 2016). The
collagen and dermal thickness, lack of resilience, enhanced SASP released from SEN fibroblasts is a typical DDR and can
DNA damage, and the induction and accumulation of SEN occur either transiently as in wound healing and develop-
fibroblasts in the skin, bone, and muscle leading to skin ment or persistently as in aging (Campisi and d’Adda di
atrophy, osteoporosis, and sarcopenia (Treiber et al., 2011). Fagagna, 2007; Rodier et al., 2009). The SASP differs
These data show that mitochondrial dysfunction and depending on the studied cell type, tissue, overall context,
enhanced superoxide anion concentrations lead to fibroblast and likely the senescence-inducing agent, although these
senescence accelerating aging phenotype in connective tis- differences have not been systematically studied (Demaria
sue‒rich organs and reduce the life span of fibroblast-specific et al., 2014; Ghosh and Capell, 2016; Meyer et al., 2017;
SOD2-deficient mice. Of note, the transcription factor Nrf2, Muñoz-Espı́n et al., 2013; Rodier et al., 2009). Recently,
which is a key regulator of the antioxidant system if overex- Basisty et al. (2020) reported a SASP Atlas. This represents a
pressed in murine skin fibroblasts, induces skin aging comprehensive proteomic database of soluble and exosomal
(Hiebert et al., 2018). Thus, an imbalance of ROS detoxifi- SASP factors, which are enforced by a variety of senescence
cation enforces tissue decline and aging. The activation of the inducers in skin fibroblasts. Each profile consists of hundreds
specific signaling pathways promoting the overexpression of of largely distinct proteins but also includes a subset of pro-
cell cycle kinase (CDK) inhibitor proteins such as p16, p21, teins elevated in all SASPs (Basisty et al., 2020). SASP factors

986 Journal of Investigative Dermatology (2021), Volume 141


M Wlaschek et al.
Fibroblast Senescence and Skin Aging

Table 1. Evidence for the Role of Fibroblast Senescence in Skin Aging


Experimental Evidence for a Role of Fibroblast Senescence in Skin Aging Reference

p16INK4a-positive SEN cells increase in the dermis of human skin and the number correlates with wrinkle formation and Ressler et al. (2006);
typical morphologic features of aging of the elastic fibers in human skin. Waaijer et al. (2016)
scRNA-seq of young versus old human skin reveals that aged fibroblasts show a functional decline, loss of priming into Salzer et al. (2018);
distinct subtypes, and a propensity for adipogenic traits with disturbed dermal‒epidermal interactions. Solé-Boldo et al. (2020)
In situ aged human dermal fibroblasts in human skin reveal in part a proteome profile identical to senescence-associated Waldera-Lupa et al. (2014);
changes. Waldera Lupa et al. (2015)
A proteome atlas depicts that in vitro‒induced senescence of human fibroblasts results in the secretion of factors enforcing Basisty et al. (2020);
skin aging, inflammaging, as well as matrix degradation. Coppé et al. (2010)
Fibroblast-specific genetic ablation of SOD2 enhanced the number of SEN (p16INK4a-positive) fibroblasts in the skin and Treiber et al. (2011)
other organs in a murine model, and this correlates with severe skin aging, osteoporosis, and sarcopenia.
Transplantation of human SEN fibroblasts into the skin of young NSG mice resulted in the spreading of senescence with the da Silva et al. (2019)
expression of senescence markers in the dermis 3 weeks after transplantation.
Organotypic human skin culture models constructed with human SEN fibroblasts show hallmarks of skin aging with disrupted Lämmermann et al., (2018);
epidermal differentiation and function. This can partly be reversed by plant extracts. Weinmüllner et al. (2020)
In a perforin-deficient mouse with reduced removal capacity of SEN cells by NK cells, SEN cells accumulate in various organs Ovadya et al. (2018)
and, importantly, in the dermis of the skin leading to structural changes in skin aging.
Topical treatment of human skin with rapamycin in vivo resulted in the reduction of p16INK4a-positive cells in combination Chung et al. (2019)
with a decrease in fine wrinkles, reduced sagging, and an increase in dermal volume.
Abbreviations: scRNA-seq, single-cell RNA sequencing; SEN, senescent.

released from SEN fibroblasts are responsible not only for the progression of malignant tumors, including squamous
skin aging and aging of other organs but also for a variety of cell carcinomas (reviewed in Coppé et al. [2010]; Farsam
age-related diseases (summarized in Childs et al. [2015]); for et al. [2016]; Liakou et al. [2016]; Mavrogonatou et al.
[2019]); and for the nonhealing state of wounds (Sindrilaru
et al., 2011). The SASP derived from human fibroblasts
consists, among many other factors, of increased concentra-
tions of IL-1, IL-6, IL-8, IL-18, matrix metalloproteinases
(MMPs), and a variety of chemokines (Coppé et al., 2010). In
addition, lipids (Ni et al., 2016) and micro RNA (Terlecki-
Zaniewicz et al., 2018) packaged in extracellular vesicles
direct and modulate basic biological functions such as
migration, proliferation, and senescence (da Silva et al.,
2019; Gonzalez-Meljem et al., 2018; Herranz and Gil,
2018; Malaquin et al., 2016; Weilner et al., 2013) of neigh-
boring cells (Faget et al., 2019; Lee and Schmitt, 2019), im-
mune cells (Prata et al., 2018), and other skin cells.
Interestingly, in vivo reprogramming and the transplantation
of mesenchymal stem cells (MSCs) suppress SASP factors
(Taguchi and Yamada, 2017; Vander Beken et al., 2019).
Interrelated paracrine loops of some of these factors such as
IL-1 and IL-6 have earlier been described to induce c-Jun
N-terminal kinase and AP-1 and, in consequence, matrix-
degrading metalloproteases as a response of human fibro-
blasts to UV irradiation and in part also for intrinsic aging
in vitro and in vivo in healthy humans (Fisher et al., 2009;
Schneider et al., 2017; Urbanski et al., 1990; Wlaschek et al.,
1994) and in intrinsic aging. IL-6, in fact, increases in elderly
adults and contributes to wrinkle formation in human skin
(Kim et al., 2012) and other age-related diseases (Beyer et al.,
Figure 2. Cell-autonomous and external cues determine fibroblast
2012; Cohen et al., 1997; Jenny et al., 2002) and increased
senescence and skin aging. Senescence-promoting and -suppressing
mechanisms depend on the quality control and its disturbances of cell‒cell
mortality (Lee et al., 2012). Senescence spreading from SEN
and cell‒matrix interactions, cellular location, and the release of a specific human fibroblasts to adjacent non-SEN human fibroblasts
SASP from fibroblasts. Malfunction drives and spreads fibroblasts senescence, in vitro and SEN hepatocytes to non-SEN hepatocytes in the
skin aging, likely the aging of other organs, aging-related diseases, and liver in vivo has earlier been shown (Acosta et al., 2013;
organismal aging. There is an intimate relationship between autonomous Nelson et al., 2018, 2012). It is of relevance to interrupt the
(mitochondrial dysfunction, enhanced concentrations of ROS, DNA damage, spreading of senescence and the SASP. In an independent
telomere attrition, epigenetic marks, loss of proteostasis, others) and external
recent systems biology approach employing a Boolean
(cell nonautonomous) cues. For further details, see the main manuscript.
SASP, senescence-associated secretory phenotype; SEN, senescent.
network technology, we were able to mechanistically show

www.jidonline.org 987
M Wlaschek et al.
Fibroblast Senescence and Skin Aging

that DNA damage‒mediated SASP triggering of IL-6 and IL-8 (Baker et al., 2016; Soto-Gamez and Demaria, 2017). The
is mainly relayed through NF-kB in human dermal fibro- genetic ablation of SEN cells was an encouraging proof for
blasts. This computational study also identified and experi- the causal role of SEN cells in aging-associated disorders.
mentally verified NF-kB essential modulator as a promising The next step was to screen for small pharmacologically
inhibitor among other inhibitors (Meyer et al., 2017). active molecules. Employing bioinformatics and small
interfering RNA technology, anti-apoptotic pathways active
SEN fibroblasts with an aberrant SASP disrupt dermal‒ in SEN cells were uncovered as senolytic targets, including
epidermal interactions. Recently, Liu et al., (2019) made the anti-apoptotic proteins BCL-2, BCL-W, and BCL-XL; the
the seminal observation that weakening of collagen type transcription factors p53 and p21; HIF1a; phosphatidyli-
XVII, which serves to anchor murine epidermal progenitor nositol-4,5-bisphosphate 3-kinase and protein kinase B; the
cells in the hemidesmosome and, in consequence, within the serine protein inhibitors (serpins); and the heat shock pro-
basement membrane of the skin, results in being replaced by tein 90, among others (Fuhrmann-Stroissnigg et al., 2017;
another progenitor cell that produces intact collagen XVII Zhu et al., 2015). With this advanced understanding, the
(Liu et al., 2019). Enhanced disintegrin and metalloproteinase rational design of drugs became possible. In this context,
and MMPs released by SEN fibroblasts in the papillary dermis navitoclax (ABT-263) is a specific inhibitor of BCL-2, BCL-
or shedding of collagen XVII induced by the SASP factor IL- XL, and BCL-W, enforcing the killing of SEN human WI-38
1b released by SEN fibroblasts (Franzke et al., 2002) may fibroblasts, human IMR-90 lung fibroblasts, and many other
contribute to the reduction of collagen XVII, with possibly cell types. Interestingly, this senolytic action is independent
constant replacement of epidermal progenitor cells, and this of the senescence-inducing trigger. Navitoclax is endowed
most likely will contribute to epidermal SC exhaustion. In with the capacity to rejuvenate SEN hematopoietic stem
addition, IGF-1 mainly released by dermal fibroblasts is cells and SEN muscle stem cells (Chang et al., 2016; Kim
essential for MSC niches and the balanced regulation of et al., 2017). Importantly, the BCL-2 inhibitor ABT-737
epidermal cell proliferation and differentiation. Keratinocytes reduced the population of SEN stem cells in the epidermis
themselves cannot synthesize IGF-1 but do express the IGF-1 in a mouse model of inducible senescence in the basal layer
receptor. IGF-1 belongs to the SASP factors, and the IGF-1 of the epidermis of the skin (Yosef et al., 2016). Currently,
released from SEN fibroblasts is significantly reduced. This the combination of two senolytic agents (such as the
is due to enhanced superoxide anion production in the tyrosinase inhibitor dasatinib and quercetin, a singlet
mitochondria of human and murine dermal fibroblasts oxygen-quenching agent) has entered clinical trials. One
in vitro and in vivo (Singh et al., 2015). In fact, earlier, we study is successfully completed now for primary pulmonary
could show that superoxide anions at increased concentra- fibrosis, a severe lung disease with a proven accumulation
tions in murine and human skin fibroblasts suppress IGF-1 of SEN cells and life-threatening fibrosis destroying the lung
signaling at the level of decreased IGF-1 receptor activation structure (Justice et al., 2019). The other disease treated with
(Singh et al., 2015). This deprived IGF-1 release, and the a senolytic approach in a clinical trial is human diabetic
inhibition of relaying the IGF-1 signal suppresses collagen kidney disease (Hickson et al., 2019). Of note, the com-
synthesis in the dermis and led to epidermal atrophy with bined therapy with quercetin and dasatinib resulted in the
enhanced accumulation of the DNA damage‒induced suppression of the low-grade inflammation with the reduc-
phosphorylated histone protein, gH2AX, and p16INK4A-posi- tion of CD68þ macrophages in the subcutaneous fat tissue
tive epidermal cells. The essential role of IGF-1 in human beneath the dermis in human individuals (Ellison-Hughes,
skin, muscle, and bone was earlier reported (Anisimov and 2020). Of note, a plant extract of goldenrod displayed a
Bartke, 2013; Gallagher and LeRoith, 2011). Reduced IGF- moderate senolytic activity and a substantial suppression of the
1 expression in the dermis of elderly humans and reduced SASP with some rejuvenating effects on human skin equivalents
IGF-1 serum concentrations directly correlate with tissue (Lämmermann et al., 2018). Collectively, senolytics hold sub-
decline in human skin aging and osteoporosis (Hassan et al., stantial promise for the successful treatment and delay of a
2015; Lamberts et al., 1997). Of note, the age of human in- variety of aging-related diseases that are due to SASP-induced
dividuals from whom dermal human fibroblasts have been low-grade inflammation.
isolated impacts on keratinocyte differentiation and wound
healing (Hausmann et al., 2019). These data clearly show Fibroblast heterogeneity and its potential role in aging
that SEN and aged fibroblasts play a major role in dermal‒ By contrast to the well-established lineage hierarchy in the
epidermal interactions and overall skin tissue decline. hematopoietic system (Zhang et al., 2018), the existence of a
hierarchy of fibroblast lineage is largely unresolved. Single-
The development of senolytic agents. Employing a genetic cell RNA sequencing (scRNA-seq) analysis recently uncov-
approach that selectively induced apoptosis of p16INK4A- ered an enormous variety of fibroblast subsets, each with a
positive SEN cells in murine models of aging-related diseases typical and reproducible transcriptional fingerprint (Driskell
led to impressive improvement (Baker et al., 2011), in and Watt, 2015; Jahoda and Gilmore, 2016; Korosec et al.,
particular of osteoarthritis, atherosclerosis, cataract, sarco- 2019; LeBleu and Neilson, 2020; Lynch and Watt, 2018;
penia, and dementia (summarized in Childs et al. [2017]). Philippeos et al., 2018; Sacco et al., 2019; Sahai et al., 2020;
Currently, it is unknown whether the genetic depletion of Salzer et al., 2018). Accordingly, a lineage hierarchy of these
SEN cells from the dermis would delay aging. The depletion newly described fibroblast subsets is slowly emerging, and it
of p16INK4A SEN cells significantly improves organ function is possible—although currently not understood—that distinct
and thus extends both health span and overall life span fibroblast subsets are more prone to senescence than others.

988 Journal of Investigative Dermatology (2021), Volume 141


M Wlaschek et al.
Fibroblast Senescence and Skin Aging

Using transplantation assays and lineage tracing, Driskell and elastin among many others (Bjorksten and Tenhu, 1990) af-
Watt (2015, 2013) showed that murine skin fibroblasts arise fects the physicomechanical properties (stiffness) of ECM
from two distinct embryonic lineages. Similarly, Jiang et al. proteins, and this likely is responsible for disrupted mecha-
(2018) characterized embryonic fibroblast lineages on the nosensing causing cell and tissue dysfunction and aging
basis of their history of expressing the EN-1 gene. EN-1 is a (Segel et al., 2019; Wang et al., 2019). Earlier, Sun et al.
transcription factor expressed in a small subset of early em- (2011) placed MSCs from old mice on the ECM from young
bryonic cells and switched off permanently later in embryo- mice and observed an impressive 16-fold increase in their
genesis (Rinkevich et al., 2015). Single-cell fate mapping, life proliferation. Although it is unclear whether enhanced stiff-
three-dimensional confocal imaging showed that wound ness of the ECM from old mice is responsible for this, reju-
healing and regeneration are driven by those fibroblast de- venated MSCs showed a decrease in intracellular ROS
scendants originating from cells that during embryogenesis concentrations and increased adenosine triphosphate levels.
did not express the EN-1 gene (engrailed history-naive cells). Interestingly, aging-dependent collagen fragmentation by
By contrast, EN-1 history‒positive fibroblasts deriving from enhanced activity of distinct MMPs impaired fibroblast‒ECM
embryonic cells, which transiently express EN-1 during interactions with reduced mechanical forces, resulting in
embryogenesis, are responsible for scarring. Transplantation fibroblast reprogramming with dampened collagen synthesis
of EN-1‒naive cells prevents scarring. Interestingly, using and enhanced proteolysis amplifying a vicious cycle of tissue
scRNA-seq of normal human skin, Vorstandlechner et al. decline (Fisher et al., 2009). Injection of hyaluronic acid
(2020) made the seminal observation that the tran- fillers into the human skin of elderly adults helped to restore
scriptomic heterogeneity of fibroblast subsets in the adult the functional capacity of aged human fibroblasts in the skin
human skin is more extended than previously anticipated in with increased collagen synthesis (Quan et al., 2013). These
the earlier described two lineage models during embryo- data may imply that the quality of the ECM impacts fibroblast
genesis. Studies based on scRNA-seq of skin from photo- function and senescence.
protected young and old human individuals (Dubois et al.,
2020; Solé-Boldo et al., 2020) and from mice (Salzer et al., Position stability and depletion of fibroblast may drive skin
2018) highlight the age-related loss of fibroblast cellular aging
identity with alteration in functional and spatial gene Tracking the same skin fibroblasts in live mice, Marsh et al.
expression signatures. Using population and single-cell (2018) show that fibroblast position is stable over time and
transcriptomics as well as long-term lineage tracing, Beni- that this stability is maintained also in the case that neigh-
tah’s group (Salzer et al., 2018) convincingly demonstrates boring fibroblasts are experimentally depleted by laser abla-
that the identity of old fibroblasts becomes undefined in a tion. Interestingly, fibroblast membranes are apparently
murine model where fibroblast signatures present in young highly dynamic in skin maintenance and homeostasis. This
skin are no longer clearly demarcated in old skin. In addition, also holds true during aging, with clustered fibroblast
old fibroblasts not only reduce the expression of genes depletion in aging skin (Marsh et al., 2018). Fibroblast
involved in the formation of the ECM but also gain adipo- membranes surprisingly spread and extend to fill the space of
genic traits. These potential tissue devastating changes can, lost adjacent fibroblasts. This extension of cell membranes
however, in part be restored by long-term caloric restriction. occurs in a Rac1-dependent manner. These data imply that
By contrast, a high-fat diet potentiates them. Apparently, positional stability and cell occupancy prime events that are
tissue decline and aging can also occur owing to the loss of key for the homeostasis of skin fibroblasts in vivo throughout
fibroblast identity and the adoption of adipogenic traits, and the lifespan of mice. This network of fibroblasts may also
this can be reversed by metabolic changes with caloric re- provide interaction substrates for recruited immune cells and,
striction. This exciting observation supports the beneficial in addition, may facilitate paracrine communication between
effects of caloric restriction, which in part has already been both immediate and more distant neighbors (Adler et al.,
translated for clinical application (Longo et al., 2015). 2018; Inaba et al., 2015; Sanders et al., 2013; Zhou et al.,
2018) and if disrupted—possibly by the release of a persis-
Fibroblast plasticity and cues from the ECM tent and proinflammatory SASP—may even drive skin aging.
There is growing evidence that fibroblasts are not only The capacity of fibroblasts to spread their membranes may
regulated by their developmental origin and position with imply that changing the cell size requires less energy
retained memory and transcriptomic signature indicating compared with the energy demand for proliferation and
where they come from (Sacco et al., 2019). In addition, their migration but is sufficient to survey putative ECM zones
morphologic and functional plasticity is further amplified by (Abdel-Haleem et al., 2017; Zanotelli et al., 2018). Interest-
epigenetic marks, by transitions from distinct cells to a ingly, although the number of SEN cells accumulates in hu-
fibroblast phenotype, by cell‒cell interactions, and impor- man skin during aging (Ressler et al., 2006), there is evidence
tantly, by the ECM itself (for review, see Fedintsev and that older human individuals when compared with young
Moskalev [2020]; LeBleu and Neilson [2020]). In fact, there individuals had fewer fibroblasts and with a greater number
is emerging evidence that stochastic nonenzymatic modifi- of longer membrane projections (Andrew et al., 1964e1965;
cation of the long-lived ECM molecules and other mac- Novotny and Gnoth, 1991; Salzer et al., 2018). The cellular
romolecules have a previously underestimated impact on depletion does not occur uniformly, but there are clustered
skin, tissue, and organ aging (Fedintsev and Moskalev, 2020). cell losses. Collectively, depletion of fibroblasts during aging
Postulated first by Björksten in 1942, this hypothesis predicts with the concomitant accumulation of SEN fibroblasts con-
that crosslinking of macromolecules such as collagen and tributes to skin aging.

www.jidonline.org 989
M Wlaschek et al.
Fibroblast Senescence and Skin Aging

Perspectives Campisi J, Kapahi P, Lithgow GJ, Melov S, Newman JC, Verdin E. From dis-
The unique opportunity to unveil the role of fibroblast subsets coveries in ageing research to therapeutics for healthy ageing. Nature
2019;571:183e92.
and modifications of various ECM proteins during skin aging
Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen
will pave the way for future research directions and the to good cells. Nat Rev Mol Cell Biol 2007;8:729e40.
development of novel preventive and therapeutic strategies. Catalgol B, Grune T. Protein pool maintenance during oxidative stress. Curr
These are currently very dynamic research foci of aging in Pharm Des 2009;15:3043e51.
general, and preventive and therapeutic strategies are Chang J, Wang Y, Shao L, Laberge RM, Demaria M, Campisi J, et al. Clearance
recently summarized in several reviews (Childs et al., 2017; of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in
Choi et al., 2011; Fedintsev and Moskalev, 2020; Prata et al., mice. Nat Med 2016;22:78e83.
2018; Toutfaire et al., 2017; Velarde and Demaria, 2016; Zhu Childs BG, Durik M, Baker DJ, van Deursen JM. Cellular senescence in aging
and age-related disease: from mechanisms to therapy. Nat Med 2015;21:
et al., 2015). 1424e35.
ORCIDs Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J,
Meinhard Wlaschek: http://orcid.org/0000-0001-5821-1140 et al. Senescent cells: an emerging target for diseases of ageing. Nat Rev
Pallab Maity: http://orcid.org/0000-0001-9769-3533 Drug Discov 2017;16:718e35.
Evgenia Makrantonaki: http://orcid.org/0000-0001-8522-2128 Choi HR, Cho KA, Kang HT, Lee JB, Kaeberlein M, Suh Y, et al. Restoration of
Karin Scharffetter-Kochanek: http://orcid.org/0000-0002-9655-685X senescent human diploid fibroblasts by modulation of the extracellular
matrix. Aging Cell 2011;10:148e57.
CONFLICT OF INTEREST
The authors state no conflict of interest. Chondrogianni N, Gonos ES. Proteasome function determines cellular ho-
meostasis and the rate of aging. Adv Exp Med Biol 2010;694:38e46.
AUTHOR CONTRIBUTIONS Chung CL, Lawrence I, Hoffman M, Elgindi D, Nadhan K, Potnis M, et al.
Conceptualization: MW, PM, KSK; Writing - Original Draft Preparation: MW, Topical rapamycin reduces markers of senescence and aging in human skin:
KSK; Writing - Review and Editing: MW, KSK, PM, EM an exploratory, prospective, randomized trial. Geroscience 2019;41:861e9.
Cohen HJ, Pieper CF, Harris T, Rao KM, Currie MS. The association of plasma
IL-6 levels with functional disability in community-dwelling elderly.
REFERENCES J Gerontol A Biol Sci Med Sci 1997;52:M201e8.
Abdel-Haleem AM, Lewis NE, Jamshidi N, Mineta K, Gao X, Gojobori T. The Coppé JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated
emerging facets of non-cancerous Warburg effect. Front Endocrinol (Lau- secretory phenotype: the dark side of tumor suppression. Annu Rev
sanne) 2017;8:279. Pathol 2010;5:99e118.
Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, et al. da Silva PFL, Ogrodnik M, Kucheryavenko O, Glibert J, Miwa S, Cameron K,
A complex secretory program orchestrated by the inflammasome controls et al. The bystander effect contributes to the accumulation of senescent
paracrine senescence. Nat Cell Biol 2013;15:978e90. cells in vivo. Aging Cell 2019;18:e12848.
Adler M, Mayo A, Zhou X, Franklin RA, Jacox JB, Medzhitov R, et al. Endo- da Silva PFL, Schumacher B. DNA damage responses in ageing. Open Biol
cytosis as a stabilizing mechanism for tissue homeostasis. Proc Natl Acad 2019;9:190168.
Sci USA 2018;115:E1926e35.
d’Adda di Fagagna F. Living on a break: cellular senescence as a DNA-
Andrew W, Behnke RH, Sato T. Changes with advancing age in the cell damage response. Nat Rev Cancer 2008;8:512e22.
population of human dermis. Gerontologia 1964e1965;10:1e19.
Demaria M, Desprez PY, Campisi J, Velarde MC. Cell autonomous and non-
Anisimov VN, Bartke A. The key role of growth hormoneeinsulineIGF-1 autonomous effects of senescent cells in the skin. J Invest Dermatol
signaling in aging and cancer. Crit Rev Oncol Hematol 2013;87:201e23. 2015;135:1722e6.
Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, et al. Naturally Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, et al.
occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature An essential role for senescent cells in optimal wound healing through
2016;530:184e9. secretion of PDGF-AA. Dev Cell 2014;31:722e33.
Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, et al. A biomarker
et al. Clearance of p16Ink4a-positive senescent cells delays ageing- that identifies senescent human cells in culture and in aging skin in vivo.
associated disorders. Nature 2011;479:232e6. Proc Natl Acad Sci USA 1995;92:9363e7.
Basisty N, Kale A, Jeon OH, Kuehnemann C, Payne T, Rao C, et al. Driskell RR, Lichtenberger BM, Hoste E, Kretzschmar K, Simons BD,
A proteomic atlas of senescence-associated secretomes for aging biomarker Charalambous M, et al. Distinct fibroblast lineages determine dermal
development. PLoS Biol 2020;18:e3000599. architecture in skin development and repair. Nature 2013;504:
Berneburg M, Gattermann N, Stege H, Grewe M, Vogelsang K, Ruzicka T, 277e81.
et al. Chronically ultraviolet-exposed human skin shows a higher mutation Driskell RR, Watt FM. Understanding fibroblast heterogeneity in the skin.
frequency of mitochondrial DNA as compared to unexposed skin and the Trends Cell Biol 2015;25:92e9.
hematopoietic system. Photochem Photobiol 1997;66:271e5.
Dubois A, Gopee N, Olabi B, Haniffa M. Defining the skin cellular com-
Beyer I, Mets T, Bautmans I. Chronic low-grade inflammation and age-related munity using single-cell genomics to advance precision medicine [e-pub
sarcopenia. Curr Opin Clin Nutr Metab Care 2012;15:12e22. ahead of print]. J Invest Dermatol 2020. https://doi.org/10.1016/j.jid.
Birch J, Barnes PJ, Passos JF. Mitochondria, telomeres and cell senescence: 2020.05.104 (accessed 25 January 2021).
implications for lung ageing and disease. Pharmacol Ther 2018;183: Eckhart L, Tschachler E, Gruber F. Autophagic control of skin aging. Front Cell
34e49. Dev Biol 2019;7:143.
Birch-Machin MA, Tindall M, Turner R, Haldane F, Rees JL. Mitochondrial Ellison-Hughes GM. First evidence that senolytics are effective at decreasing
DNA deletions in human skin reflect photo- rather than chronologic aging. senescent cells in humans. EBioMedicine 2020;56:102473.
J Invest Dermatol 1998;110:149e52.
Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent
Bjorksten J, Tenhu H. The crosslinking theory of aging — added evidence. Exp effects of SASP in cancer. Nat Rev Cancer 2019;19:439e53.
Gerontol 1990;25:91e5.
Farsam V, Basu A, Gatzka M, Treiber N, Schneider LA, Mulaw MA, et al.
Bodnar AG, Ouellette M, Frolkis M, Holt SE, Chiu CP, Morin GB, et al. Senescent fibroblast-derived chemerin promotes squamous cell carcinoma
Extension of life-span by introduction of telomerase into normal human migration. Oncotarget 2016;7:83554e69.
cells. Science 1998;279:349e52.
Fedintsev A, Moskalev A. Stochastic non-enzymatic modification of long-
Campisi J. Senescent cells, tumor suppression, and organismal aging: good lived macromolecules - a missing hallmark of aging. Ageing Res Rev
citizens, bad neighbors. Cell 2005;120:513e22. 2020;62:101097.

990 Journal of Investigative Dermatology (2021), Volume 141


M Wlaschek et al.
Fibroblast Senescence and Skin Aging

Fisher GJ, Quan T, Purohit T, Shao Y, Cho MK, He T, et al. Collagen frag- Krishnamurthy J, Torrice C, Ramsey MR, Kovalev GI, Al-Regaiey K, Su L, et al.
mentation promotes oxidative stress and elevates matrix metalloproteinase- Ink4a/Arf expression is a biomarker of aging. J Clin Invest 2004;114:
1 in fibroblasts in aged human skin. Am J Pathol 2009;174:101e14. 1299e307.
Franzke CW, Tasanen K, Schäcke H, Zhou Z, Tryggvason K, Mauch C, et al. Krutmann J, Schroeder P. Role of mitochondria in photoaging of human skin:
Transmembrane collagen XVII, an epithelial adhesion protein, is shed from the defective powerhouse model. J Investig Dermatol Symp Proc 2009;14:
the cell surface by ADAMs. EMBO J 2002;21:5026e35. 44e9.
Fuhrmann-Stroissnigg H, Ling YY, Zhao J, McGowan SJ, Zhu Y, Brooks RW, Lamberts SW, van den Beld AW, van der Lely AJ. The endocrinology of aging.
et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Science 1997;278:419e24.
Commun 2017;8:422. Lämmermann I, Terlecki-Zaniewicz L, Weinmüllner R, Schosserer M,
Gallagher EJ, LeRoith D. Is growth hormone resistance/IGF-1 reduction good Dellago H, de Matos Branco AD, et al. Blocking negative effects of
for you? Cell Metab 2011;13:355e6. senescence in human skin fibroblasts with a plant extract. NPJ Aging Mech
Dis 2018;11(4):4.
Ghosh K, Capell BC. The senescence-associated secretory phenotype:
critical effector in skin cancer and aging. J Invest Dermatol 2016;136: LeBleu VS, Neilson EG. Origin and functional heterogeneity of fibroblasts.
2133e9. FASEB J 2020;34:3519e36.
Gonzalez-Meljem JM, Apps JR, Fraser HC, Martinez-Barbera JP. Paracrine Lee JK, Bettencourt R, Brenner D, Le TA, Barrett-Connor E, Loomba R. As-
roles of cellular senescence in promoting tumourigenesis. Br J Cancer sociation between serum interleukin-6 concentrations and mortality in
2018;118:1283e8. older adults: the Rancho Bernardo study. PLoS One 2012;7:e34218.
Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Liakou E, Mavrogonatou E, Pratsinis H, Rizou S, Evangelou K, Panagiotou PN,
Cellular senescence: defining a path forward. Cell 2019;179:813e27. et al. Ionizing radiation-mediated premature senescence and paracrine in-
teractions with cancer cells enhance the expression of syndecan 1 in human
Gu Y, Han J, Jiang C, Zhang Y. Biomarkers, oxidative stress and autophagy in
breast stromal fibroblasts: the role of TGF-b. Aging 2016;8:1650e69.
skin aging. Ageing Res Rev 2020;59:101036.
Lee S, Schmitt CA. The dynamic nature of senescence in cancer. Nat Cell Biol
Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of
2019;21:94e101.
human fibroblasts. Nature 1990;345:458e60.
Liu N, Matsumura H, Kato T, Ichinose S, Takada A, Namiki T, et al. Stem cell
Hassan AM, Elgarhy LH, Mohammad WS, Farag NE. Relation of insulin-like
competition orchestrates skin homeostasis and ageing. Nature 2019;568:
growth factor to skin ageing. J Egypt Women Dermatol Soc 2015;12:
344e50.
174e9.
Longo VD, Antebi A, Bartke A, Barzilai N, Brown-Borg HM, Caruso C, et al.
Hausmann C, Zoschke C, Wolff C, Darvin ME, Sochorová M, Ková cik A, et al.
Interventions to slow aging in humans: are we ready? Aging Cell 2015;14:
Fibroblast origin shapes tissue homeostasis, epidermal differentiation, and
497e510.
drug uptake. Sci Rep 2019;9:2913.
Lozano-Torres B, Estepa-Fernández A, Rovira M, Orzáez M, Serrano M,
Herranz N, Gil J. Mechanisms and functions of cellular senescence. J Clin
Martı́nez-Máñez R, et al. The chemistry of senescence. Nat Rev Chem
Invest 2018;128:1238e46.
2019;3:426e41.
Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK,
Lynch MD, Watt FM. Fibroblast heterogeneity: implications for human dis-
et al. Senolytics decrease senescent cells in humans: preliminary report
ease. J Clin Invest 2018;128:26e35.
from a clinical trial of dasatinib plus quercetin in individuals with diabetic
kidney disease. EBioMedicine 2019;47:446e56. Malaquin N, Martinez A, Rodier F. Keeping the senescence secretome under
control: molecular reins on the senescence-associated secretory pheno-
Hiebert P, Wietecha MS, Cangkrama M, Haertel E, Mavrogonatou E,
type. Exp Gerontol 2016;82:39e49.
Stumpe M, et al. Nrf2-mediated fibroblast reprogramming drives
cellular senescence by targeting the matrisome. Dev Cell 2018;46: Marsh E, Gonzalez DG, Lathrop EA, Boucher J, Greco V. Positional stability
145e61.e10. and membrane occupancy define skin fibroblast homeostasis in vivo. Cell
2018;175:1620e33.e13.
Hoeijmakers JHJ. DNA damage, aging, and cancer. N Engl J Med 2009;361:
1475e85. Mavrogonatou E, Pratsinis H, Papadopoulou A, Karamanos NK, Kletsas D.
Extracellular matrix alterations in senescent cells and their significance in
Inaba M, Buszczak M, Yamashita YM. Nanotubes mediate nicheestem-cell
tissue homeostasis. Matrix Biol 2019;75e76:27e42.
signalling in the Drosophila testis. Nature 2015;523:329e32.
Meyer P, Maity P, Burkovski A, Schwab J, Müssel C, Singh K, et al. A model of
Jahoda CAB, Gilmore AC. What lies beneath: Wnt/b-catenin signaling and
the onset of the senescence associated secretory phenotype after DNA
cell fate in the lower dermis. J Invest Dermatol 2016;136:1084e7.
damage induced senescence. PLoS Comp Biol 2017;13:e1005741.
Jenny NS, Tracy RP, Ogg MS, Luong le A, Kuller LH, Arnold AM, et al. In the
Muñoz-Espı́n D, Cañamero M, Maraver A, Gómez-López G, Contreras J,
elderly, interleukin-6 plasma levels and the 174G>C polymorphism are
Murillo-Cuesta S, et al. Programmed cell senescence during mammalian
associated with the development of cardiovascular disease. Arterioscler
embryonic development. Cell 2013;155:1104e18.
Thromb Vasc Biol 2002;22:2066e71.
Muñoz-Espı́n D, Serrano M. Cellular senescence: from physiology to pa-
Jiang D, Correa-Gallegos D, Christ S, Stefanska A, Liu J, Ramesh P, et al.
thology. Nat Rev Mol Cell Biol 2014;15:482e96.
Two succeeding fibroblastic lineages drive dermal development and
the transition from regeneration to scarring. Nat Cell Biol 2018;20: Nelson G, Kucheryavenko O, Wordsworth J, von Zglinicki T. The senescent
422e31. bystander effect is caused by ROS-activated NF-kB signaling. Mech Ageing
Dev 2018;170:30e6.
Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK,
et al. Senolytics in idiopathic pulmonary fibrosis: results from a first-in- Nelson G, Wordsworth J, Wang C, Jurk D, Lawless C, Martin-Ruiz C, et al.
human, open-label, pilot study. EBioMedicine 2019;40:554e63. A senescent cell bystander effect: senescence-induced senescence. Aging
Cell 2012;11:345e9.
Kim HN, Chang J, Shao L, Han L, Iyer S, Manolagas SC, et al. DNA damage
and senescence in osteoprogenitors expressing Osx1 may cause their Ni C, Narzt MS, Nagelreiter IM, Zhang CF, Larue L, Rossiter H, et al. Auto-
decrease with age. Aging Cell 2017;16:693e703. phagy deficient melanocytes display a senescence associated secretory
phenotype that includes oxidized lipid mediators. Int J Biochem Cell Biol
Kim OY, Chae JS, Paik JK, Seo HS, Jang Y, Cavaillon JM, et al. Effects of aging
2016;81:375e82.
and menopause on serum interleukin-6 levels and peripheral blood
mononuclear cell cytokine production in healthy nonobese women. Age Novotny GE, Gnoth C. Variability of fibroblast morphology in vivo: a silver
2012;34:415e25. impregnation study on human digital dermis and subcutis. J Anat
1991;177:195e207.
Korosec A, Frech S, Gesslbauer B, Vierhapper M, Radtke C, Petzelbauer P,
et al. Lineage identity and location within the dermis determine the func- Ovadya Y, Landsberger T, Leins H, Vadai E, Gal H, Biran A, et al. Impaired
tion of papillary and reticular fibroblasts in human skin. J Invest Dermatol immune surveillance accelerates accumulation of senescent cells and ag-
2019;139:342e51. ing. Nat Commun 2018;9:5435.

www.jidonline.org 991
M Wlaschek et al.
Fibroblast Senescence and Skin Aging

Passos JF, Saretzki G, von Zglinicki T. DNA damage in telomeres and mito- Treiber N, Maity P, Singh K, Kohn M, Keist AF, Ferchiu F, et al. Accelerated aging
chondria during cellular senescence: is there a connection? Nucleic Acids phenotype in mice with conditional deficiency for mitochondrial superoxide
Res 2007;35:7505e13. dismutase in the connective tissue. Aging Cell 2011;10:239e54.
Philippeos C, Telerman SB, Oulès B, Pisco AO, Shaw TJ, Elgueta R, et al. Tuttle CSL, Waaijer MEC, Slee-Valentijn MS, Stijnen T, Westendorp R,
Spatial and single-cell transcriptional profiling identifies functionally Maier AB. Cellular senescence and chronological age in various human
distinct human dermal fibroblast subpopulations. J Invest Dermatol tissues: a systematic review and meta-analysis. Aging Cell 2020;19:e13083.
2018;138:811e25. Urbanski A, Schwarz T, Neuner P, Krutmann J, Kirnbauer R, Köck A, et al.
Prata LGPL, Ovsyannikova IG, Tchkonia T, Kirkland JL. Senescent cell Ultraviolet light induces increased circulating interleukin-6 in humans.
clearance by the immune system: emerging therapeutic opportunities. J Invest Dermatol 1990;94:808e11.
Semin Immunol 2018;40:101275. Vander Beken S, de Vries JC, Meier-Schiesser B, Meyer P, Jiang D,
Quan T, Wang F, Shao Y, Rittié L, Xia W, Orringer JS, et al. Enhancing Sindrilaru A, et al. Newly defined ATP-binding cassette subfamily B
structural support of the dermal microenvironment activates fibroblasts, Member 5 positive dermal mesenchymal stem cells promote healing of
endothelial cells, and keratinocytes in aged human skin in vivo. J Invest chronic iron-overload wounds via secretion of interleukin-1 receptor
Dermatol 2013;133:658e67. antagonist. Stem Cells 2019;37:1057e74.
Ressler S, Bartkova J, Niederegger H, Bartek J, Scharffetter-Kochanek K, Jan- Velarde MC, Demaria M. Targeting senescent cells: possible implications for
sen-Dürr P, et al. p16 INK4A is a robust in vivo biomarker of cellular aging delaying skin aging: a mini-review. Gerontology 2016;62:513e8.
in human skin. Aging Cell 2006;5:379e89. Vorstandlechner V, Laggner M, Kalinina P, Haslik W, Radtke C, Shaw L, et al.
Rinkevich Y, Walmsley GG, Hu MS, Maan ZN, Newman AM, Drukker M, Deciphering the functional heterogeneity of skin fibroblasts using single-
et al. Skin fibrosis. Identification and isolation of a dermal lineage with cell RNA sequencing. FASEB J 2020;34:3677e92.
intrinsic fibrogenic potential. Science 2015;348:aaa2151. Waaijer ME, Gunn DA, Adams PD, Pawlikowski JS, Griffiths CE, van
Rinnerthaler M, Bischof J, Streubel MK, Trost A, Richter K. Oxidative stress in Heemst D, et al. P16INK4a positive cells in human skin are indicative of
aging human skin. Biomolecules 2015;5:545e89. local elastic fiber morphology, facial wrinkling, and perceived age.
Rodier F, Coppé JP, Patil CK, Hoeijmakers WA, Muñoz DP, Raza SR, et al. J Gerontol A Biol Sci Med Sci 2016;71:1022e8.
Persistent DNA damage signalling triggers senescence-associated inflam- Waldera Lupa DM, Kalfalah F, Safferling K, Boukamp P, Poschmann G,
matory cytokine secretion [published correction appears in Nat Cell Biol Volpi E, et al. Characterization of skin aging-associated secreted proteins
2009 Oct;11:1272]. Nat Cell Biol 2009;11:973e9. (SAASP) produced by dermal fibroblasts isolated from intrinsically aged
Sacco AM, Belviso I, Romano V, Carfora A, Schonauer F, Nurzynska D, et al. human skin. J Invest Dermatol 2015;135:1954e68.
Diversity of dermal fibroblasts as major determinant of variability in cell Waldera-Lupa DM, Kalfalah F, Florea AM, Sass S, Kruse F, Rieder V,
reprogramming. J Cell Mol Med 2019;23:4256e68. et al. Proteome-wide analysis reveals an age-associated cellular
Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. phenotype of in situ aged human fibroblasts. Aging (Albany NY)
A framework for advancing our understanding of cancer-associated fibro- 2014;6:856e78.
blasts. Nat Rev Cancer 2020;20:174e86. Wang AS, Dreesen O. Biomarkers of cellular senescence and skin aging.
Salzer MC, Lafzi A, Berenguer-Llergo A, Youssif C, Castellanos A, Solanas G, Front Genet 2018;9:247.
et al. Identity noise and adipogenic traits characterize dermal fibroblast. Wang W, Lollis EM, Bordeleau F, Reinhart-King CA. Matrix stiffness regulates
Aging Cell 2018;175:1575e90.e22. vascular integrity through focal adhesion kinase activity. FASEB J 2019;33:
Sanders TA, Llagostera E, Barna M. Specialized filopodia direct long-range 1199e208.
transport of SHH during vertebrate tissue patterning. Nature 2013;497: Weilner S, Schraml E, Redl H, Grillari-Voglauer R, Grillari J. Secretion of
628e32. microvesicular miRNAs in cellular and organismal aging. Exp Gerontol
Schneider LA, Raizner K, Wlaschek M, Brenneisen P, Gethöffer K, Scharf- 2013;48:626e33.
fetter-Kochanek K. UVA-1 exposure in vivo leads to an IL-6 surge within the Weinmüllner R, Zbiral B, Becirovic A, Stelzer EM, Nagelreiter F,
skin. Exp Dermatol 2017;26:830e2. Schosserer M, et al. Organotypic human skin culture models constructed
Segel M, Neumann B, Hill MFE, Weber IP, Viscomi C, Zhao C, et al. Niche with senescent fibroblasts show hallmarks of skin aging. NPJ Aging Mech
stiffness underlies the ageing of central nervous system progenitor cells Dis 2020;6:4.
[published correction appears in Nature 2019;573:E3]. Nature 2019;573: Wlaschek M, Heinen G, Poswig A, Schwarz A, Krieg T, Scharffetter-
130e4. Kochanek K. UVA-induced autocrine induction of fibroblast-derived
Sindrilaru A, Peters T, Wieschalka S, Baican C, Baican A, Peter H, et al. An collagenase/MMP-1 by interrelated loops of interleukin-1 and inter-
unrestrained proinflammatory M1 macrophage population induced by iron leukin-6. Photochem Photobiol 1994;59:550e6.
impairs wound healing in humans and mice. J Clin Invest 2011;121: Yosef R, Pilpel N, Tokarsky-Amiel R, Biran A, Ovadya Y, Cohen S, et al.
985e97. Directed elimination of senescent cells by inhibition of BCL-W and BCL-
Singh K, Maity P, Krug L, Meyer P, Treiber N, Lucas T, et al. Superoxide anion XL. Nat Commun 2016;7:11190.
radicals induce IGF -1 resistance through concomitant activation of PTP 1 Yu GL, Bradley JD, Attardi LD, Blackburn EH. In vivo alteration of telomere
B and PTEN. EMBO Mol Med 2015;7:59e77. sequences and senescence caused by mutated Tetrahymena telomerase
Solé-Boldo L, Raddatz G, Schütz S, Mallm JP, Rippe K, Lonsdorf AS, et al. RNAs. Nature 1990;344:126e32.
Single-cell transcriptomes of the human skin reveal age-related loss of Zanotelli MR, Goldblatt ZE, Miller JP, Bordeleau F, Li J, VanderBurgh JA, et al.
fibroblast priming. Commun Biol 2020;3:188. Regulation of ATP utilization during metastatic cell migration by collagen
Soto-Gamez A, Demaria M. Therapeutic interventions for aging: the case of architecture. Mol Biol Cell 2018;29:1e9.
cellular senescence. Drug Discov Today 2017;22:786e95. Zhang Y, Gao S, Xia J, Liu F. Hematopoietic hierarchy e an updated roadmap.
Sun Y, Li W, Lu Z, Chen R, Ling J, Ran Q, et al. Rescuing replication and Trends Cell Biol 2018;28:976e86.
osteogenesis of aged mesenchymal stem cells by exposure to a young Zhou X, Franklin RA, Adler M, Jacox JB, Bailis W, Shyer JA, et al. Circuit
extracellular matrix. FASEB J 2011;25:1474e85. design features of a stable two-cell system. Cell 2018;172:744e57.e17.
Taguchi J, Yamada Y. Unveiling the role of senescence-induced cellular Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, et al. The
plasticity. Cell Stem Cell 2017;20:293e4. Achilles’ heel of senescent cells: from transcriptome to senolytic drugs.
Terlecki-Zaniewicz L, Lämmermann I, Latreille J, Bobbili MR, Pils V, Aging Cell 2015;14:644e58.
Schosserer M, et al. Small extracellular vesicles and their miRNA cargo are
anti-apoptotic members of the senescence-associated secretory phenotype.
Aging 2018;10:1103e32. This work is licensed under a Creative Commons
Toutfaire M, Bauwens E, Debacq-Chainiaux F. The impact of cellular senes- Attribution-NonCommercial-NoDerivatives 4.0
cence in skin ageing: a notion of mosaic and therapeutic strategies. Bio- International License. To view a copy of this license, visit
chem Pharmacol 2017;142:1e12.
http://creativecommons.org/licenses/by-nc-nd/4.0/

992 Journal of Investigative Dermatology (2021), Volume 141

You might also like