Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Received: 20 March 2021 Revised: 10 May 2021 Accepted: 17 May 2021

DOI: 10.1111/wrr.12952

PERSPECTIVE ARTICLE

Myofibroblast fate plasticity in tissue repair and fibrosis:


Deactivation, apoptosis, senescence and reprogramming

Wolfgang Merkt MD1,2 | Yan Zhou MSc1,3 | Hongwei Han PhD1 |


David Lagares PhD1

1
Fibrosis Research Center, Center for
Immunology and Inflammatory Diseases, Abstract
Division of Pulmonary and Critical Care In response to tissue injury, fibroblasts differentiate into professional repair cells
Medicine, Massachusetts General Hospital,
Harvard Medical School, Boston, called myofibroblasts, which orchestrate many aspects of the normal tissue
Massachusetts repair programme including synthesis, deposition and contraction of extracellular
2
Department of Hematology, Oncology and
matrix proteins, leading to wound closure. Successful tissue repair responses
Rheumatology, Internal Medicine V, University
Hospital of Heidelberg, Heidelberg, Germany involve termination of myofibroblast activities in order to prevent pathologic
3
Department of Physiology, Xiangya Medical fibrotic scarring. Here, we discuss the cellular and molecular mechanisms
School, Central South University, Changsha,
China limiting myofibroblast activities during physiological tissue repair, including
myofibroblast deactivation, apoptosis, reprogramming and immune clearance of
Correspondence
David Lagares, Fibrosis Research Center, senescent myofibroblasts. In addition, we summarize pathological mechanisms
Center for Immunology and Inflammatory leading to myofibroblast persistence and survival, a hallmark of fibrotic diseases.
Diseases, Division of Pulmonary and Critical
Care Medicine, Massachusetts General Finally, we discuss emerging anti-fibrotic therapies aimed at targeting
Hospital, Harvard Medical School, Boston, MA, myofibroblast fate such as senolytics, gene therapy, cellular immunotherapy and
USA.
Email: dlagares@mgh.harvard.edu CAR-T cells.

KEYWORDS
apoptosis, cellular senescence, fate, fibrosis, myofibroblast, plasticity, reprogramming, tissue
repair

1 | I N T RO DU CT I O N : M YO F I BR O B LA S T pro-repair responses during normal wound healing, ultimately promot-


F A T E S U P O N CO M P L E T I O N OF TI S S U E ing fibroblast proliferation and differentiation into professional repair
REPAIR cells named myofibroblasts.1 Activated myofibroblasts are character-
ized by increased synthesis of ECM proteins including type I collagen
During homeostasis, scattered tissue-resident fibroblasts actively and the neo-expression of alpha smooth muscle actin (α-SMA), a pro-
maintain the fitness of connective tissues through processes of syn- tein that confers these cells a hyper-contractile phenotype.2 These
thesis, degradation and remodelling of extracellular matrix (ECM) pro- two cellular activities are essential at promoting wound closure and
teins. These processes are controlled through a series of feedback preventing blood loss and infections.3 During the late phase of tissue
mechanisms involving both mechanical and biochemical cues that repair, myofibroblasts orchestrate a series of cellular responses aimed
instruct fibroblast to produce or degrade the ECM. Upon tissue injury, at fully regenerating or partially restoring parenchyma function.1 Suc-
ECM breakdown, haemostasis and inflammation unleash fibroblast cessful tissue repair responses involve termination of myofibroblast

Abbreviations: α-SMA, α smooth muscle actin; AAV, adenoassociated virus; BET, bromodomain and extra-terminal; c-Abl, cellular Abelson; CKD, chronic kidney disease; DNMT3A, DNA
methyltransferase 3A; DRs, death receptors; ECM, Extracellular Matrix; ET, endothelin; FAP, fibroblast activation protein; FasL, Fas ligand; FGF1, fibroblast growth factor 1; H3K9, histone 3
lysine 9; H3K9Me3, histone 3 lysine 9 trimethylation; HDAC4, histone deacetylase 4; HGF, hepatic growth factor; IFNγ, Interferon gamma; IL-1β, interleukin-1β; IPF, idiopathic pulmonary
fibrosis; kPa, kilopascal; MeCP2, methyl cap binding protein 2; miRs, microRNAs; MOMP, mitochondrial outer membrane permeabilization; MRTF-A/B, myocardin-related transcription factor-A/
B; NK, natural killer; PDGF, Platelet-derived growth factor; PPARγ, peroxisome proliferator-activated receptor gamma; ROS, reactive oxygen species; SAHA, suberoylanilide hydroxamic acid;
SASP, senescence-associated secretory phenotype; SSc, systemic sclerosis; TAZ, transcriptional coactivator with PDZ-binding motif; TGFβ, transforming growth factor; TRAIL, TNF-related
apoptosis inducing ligand; uPAR, urokinase-type plasminogen activator receptor; YAP, yes-associated protein..

Wound Rep Reg. 2021;1–14. wileyonlinelibrary.com/journal/wrr © 2021 The Wound Healing Society. 1
2 MERKT ET AL.

activities in order to prevent maladaptive repair responses charac- myofibroblasts are “primed for death,” a mechanism that enables
terized by pathologic fibrotic scarring.4,5 Given the importance of rapid elimination of these cells upon completion of the tissue repair
myofibroblasts in balancing regeneration and fibrosis, a growing programme. On the molecular level, this process is controlled by
body of research is currently addressing the mechanisms regulating dynamic changes in the “mitochondrial apoptotic priming” of myo-
myofibroblast activities during the resolution phase of wound fibroblasts across the different phases of the tissue repair programme.
repair and fibrosis.6 In this review, we will summarize the state of
the art regarding cellular and molecular mechanisms limiting
myofibroblast activities during normal tissue repair, including 2.1 | Mitochondrial priming controls myofibroblast
myofibroblast deactivation, apoptosis, reprogramming and immune predisposition to apoptosis
clearance of senescent myofibroblasts (Figure 1). We will also dis-
cuss escape mechanisms leading to myofibroblast persistence and Apoptotic priming is dynamically regulated by a molecular rheostat
the development of fibrosis. Finally, we will discuss emerging thera- involving the BCL-2 family of proteins, which compromises both pro-
peutic strategies aimed at reducing myofibroblast activities to treat apoptotic and anti-apoptotic members that control the mitochondrial
fibrotic diseases. or intrinsic pathway of apoptosis.14,16 This pathway is triggered by
intracellular pro-apoptotic stimuli such as oxidative stress, DNA dam-
age or oncogene activation, ultimately inducing mitochondrial outer
2 | MYOFIBROBLAST APOPTOSIS DURING membrane permeabilization (MOMP), cytochrome C release and
W O U ND H E A L I N G activation of executioner caspases. Strong inducers of mitochondrial
apoptosis such as the BCL-2 family member BIM are highly expressed
Successful tissue repair requires timely resolution of the inflammatory in activated myofibroblasts compared to resting fibroblasts.6,17 The
and fibroproliferative responses, ultimately leading to reduction in tis- mechanisms increasing levels of BIM and other pro-apoptotic mole-
sue cellularity. Early reports published in the 1990s proposed that cules in myofibroblasts are poorly understood, although increased
myofibroblast apoptosis is a major mechanism responsible for production of mitochondrial reactive oxygen species (ROS) has been
myofibroblast clearance in skin repair, as evidenced by morphological associated with mitochondrial damage and apoptotic signalling in
changes and accumulation of DNA double-strand brakes in α-SMA+ these cells.15,18,19 Platelet-derived growth factor (PDGF) signalling has
cells.4,5,7 More recent studies have validated this concept in animal been similarly shown to prime cancer-associated myofibroblasts for
models of tissue injury, further supporting the notion that apoptosis.20 Despite being tonically primed for death, myofibroblasts
programmed cell death plays a central role in wound repair by pro- evade apoptosis by upregulating pro-survival molecules that directly
moting controlled elimination of myofibroblasts during the resolution inhibit pro-death signalling. We and others have shown that pro-
of the healing programme.7–10 Whereas the mechanisms that pro- apoptotic activities of BIM are selectively blocked by the anti-
mote fibroblast proliferation and activation into myofibroblasts have apoptotic BCL-2 family protein BCL-XL, which directly binds to and
been extensively studied and reviewed,11–13 the molecular underpin- sequesters BIM in myofibroblasts' mitochondria.17 Up-regulation of
nings that control myofibroblast apoptosis remain poorly understood. pro-survival BCL-2 proteins including BCL-XL is induced by growth
We have recently shown that differentiation of fibroblasts into acti- factors, cytokines and ECM cues. For instance, the cytokine trans-
vated myofibroblasts associates with augmented predisposition to forming growth factor beta (TGFβ), the vasoactive peptide
apoptosis in these cells,6,14,15 increasing the readiness of myo- endothelin-1 (ET-1) and PDGF have been shown to promote an
fibroblasts to activate programmed cell death. Thus, activated apoptosis-resistant phenotype in myofibroblasts by activating pro-

F I G U R E 1 Myofibroblast fate upon termination of the tissue repair programme. Following tissue injury, extracellular matrix (ECM) breakdown
and inflammatory cytokines such as TGFb promote fibroblast differentiation into activated myofibroblasts, which are characterized by increased
synthesis of ECM and the neo-expression of alpha smooth muscle actin (α-SMA), a protein that confers these cells a hyper-contractile phenotype.
These two cellular activities are essential for promoting wound closure and reepithelialization. Successful tissue repair responses involve
termination of myofibroblast activities via alternative mechanisms including deactivation, apoptosis, immune clearance of senescent
myofibroblasts by T and natural killer (NK) cells and reprograming
MERKT ET AL. 3

survival signalling pathways such as FAK-PI3K-AKT and ABL.21–27 stiffening of the ECM has been shown to activate the pro-apoptotic
Similarly, matrix cues including ECM stiffness promote myofibroblast Mdm4-p53 pathway and promote myofibroblast apoptosis.35 The cel-
evasion of apoptosis by activating survival mechanotransduction path- lular mechanisms promoting ECM degradation and softening remain
ways such as integrin-FAK-ROCK-YAP/TAZ signalling.6,12 We and poorly understood but may involve reprogramming of injury-
others have recently shown that mechanical activation of YAP/TAZ associated macrophages that switch their phenotype from pro-repair
directly regulates expression of the anti-apoptotic protein BCL-XL in to matrix-degrading phenotype during the resolution of tissue
17
primed for death myofibroblasts, ensuring their survival. In addition, repair.36–38 Accordingly, molecules involved in the degradation and
TGFβ also blocks the extrinsic pathway of apoptosis by inhibiting Fas– uptake of collagen such as uPARP and Mfge8 have shown to promote
Fas ligand (FasL) signalling, a mechanism dependent on blockade of ECM degradation and prevent tissue fibrosis in mouse models.39,40
ceramide generation by sphingomyelinase.28 Taken together,
increased mitochondrial priming in activated myofibroblasts predis-
poses these cells towards apoptosis. Primed for death myofibroblasts 2.2.2 | Growth factor withdrawal
are highly dependent on the activation of survival pathways and
expression of anti-apoptotic proteins of the BCL-2 family members. In During tissue repair, primed for death myofibroblasts become
the following section, we discuss cellular and molecular mechanisms addicted to cytokines and growth factors that activate pro-survival
that selectively inhibit these pro-survival mechanisms during the reso- signalling pathways, ensuring their survival. Levels of TGFβ, ET-1
lution phase of wound healing, ultimately leading to myofibroblast and PDGF, which are released by inflammatory cells and platelets
clearance via apoptosis. during the inflammatory phase of the healing programme, are
reduced during the resolution phase,6,41,42 putting forward the
hypothesis that withdrawal of survival signals upon completion of
2.2 | Mechanisms inducing apoptosis in “primed the tissue repair programme leads to myofibroblast apoptosis. 27,43
for death” myofibroblasts While this hypothesis has not been demonstrated in vivo, several
reports have shown that starvation of cultured myofibroblasts
2.2.1 | Matrix softening and degradation associates with reduced FAK-PI3K-AKT signalling and apoptosis.
Together, growth factor “dependence” may control myofibroblast
Upon injury, the wound is filled with a provisional matrix composed of clearance during wound healing.
fibrin, fibronectin and collagens, which matures into scar tissue during
the remodelling phase of the healing programme. As the tissue repair
programme progresses, the mechanical properties of the granulation 2.2.3 | Pro-apoptotic factors
tissue change, in particular by increasing its stiffness and resistance to
deformation. These changes in matrix stiffness have been shown Another plausible mechanism involves factors that specifically
to directly modulate the biology of fibroblasts during wound repair by inhibit survival signalling in myofibroblasts. For instance, fibroblast
promoting their mechano-activation into myofibroblasts.2,12 Previous growth factor 1 (FGF1), interleukin-1β (IL-1β) and hepatic growth
reports have shown that dermal stiffness increases from 1 kPa in factor (HGF) have been shown to deactivate the FAK-PI3K-AKT
healthy skin tissues up to 20 kPa in 7 days post-wounding and 50 kPa pathway and induce apoptosis of myofibroblasts from skin granula-
in mature granulation tissue 14 days post-injury.29 At this point, tion tissue, but not in healthy control fibroblasts. Similarly, the
wound closure is almost completed due to full activation of hyper- prostaglandin PGE2 blocks AKT signalling and increases the sensi-
contractile myofibroblasts.30 Of note, massive degradation of the tivity of myofibroblasts to Fas-induced apoptosis via the extrinsic
ECM is observed once the wound is closed, which associates with pathway. In this regard, activation of the extrinsic pathway of apo-
myofibroblast apoptosis.31 Several reports have shown that the ptosis by FasL and TNF-related apoptosis-inducing ligand (TRAIL)
degree of myofibroblasts apoptosis appears to correlate with severity has been similarly reported to promote myofibroblast apoptosis
of ECM stiffness and tension drop. For instance, 15% of activated in vivo.8,44,45 Accordingly, loss of pro-apoptotic Fas signalling in
myofibroblasts undergo apoptosis following release of anchored colla- myofibroblasts impairs homeostatic tissue repair resolution and
gen gels in vitro from high to low tension,31 whereas 40% of activated promotes pulmonary fibrosis in mice.8 In humans, bronchoalveolar
myofibroblasts activate apoptosis when tension drops from very high lavage fluid from patients in the resolution phase of lung repair,
to zero.32,33 In vivo, releasing the edges of a previously splinted exci- but not from patients with active lung injury, induces myofibroblast
sional wound, which leads to tension drop from high to medium, cau- apoptosis, further suggesting that myofibroblasts are actively and
ses apoptosis in 10% of the cells, albeit the percentage of apoptotic timely eliminated via apoptosis.46
34
myofibroblasts was not calculated. Mechanistically, ECM degrada- Together, the termination of myofibroblasts' pro-repair activities
tion and softening have been shown to promote myofibroblast apo- emerges as a tightly regulated process in which both intrinsic apopto-
ptosis by inhibiting mechanically activated FAK signalling and tic priming as well as spatiotemporal regulation of mechanical and
reducing levels of pro-survival BCL-XL in primed for the myo- biochemical factors fine-tune the activation of myofibroblast apopto-
fibroblasts in vitro and in vivo models.31,34 More recently, de- sis during the resolution phase of the repair programme.
4 MERKT ET AL.

2.2.4 | Myofibroblast senescence and immune in myofibroblast apoptosis by secreting interferon gamma (IFNγ), which
clearance facilitates myofibroblast apoptosis by NK cells in the context of liver
injury.71–73 Finally, myofibroblast apoptotic bodies are subsequently
In addition to self-clearance by apoptosis, acquisition of a senescent phagocytosed by macrophages via efferocytosis, which associates with
phenotype has been shown to reduce myofibroblast activities and anti-inflammatory and wound healing resolution responses.74,75
promote their clearance by cytotoxic immune cells. The role of cellular
senescence in normal tissue repair remains poorly understood,
although it appears to be a mechanism to arrest myofibroblasts and to 3 | MYOFIBROBLAST DEACTIVATION
limit their pro-repair activities47,48 during the resolution phase of the A ND R E P R O G RA M M I N G D U R I N G W O U N D
tissue repair programme in the liver,49 lungs,50,51 skin,52,53 kidney,54 HE AL IN G
oral submucosa55 and pancreas.56 Mechanistically, the matricellular
protein CCN1 has been shown to promote myofibroblast senescence Genetic lineage-tracing studies in mice have recently demonstrated
in vivo in the context of liver and skin injury, promoting liver regenera- that myofibroblasts undergo dynamic phenotypic transitions through
tion and limiting fibrosis in mouse models.52,57 In addition, the different activation states in vivo.10,76–78 These studies show that myo-
senescent phenotype appears to target myofibroblast for recognition fibroblasts get deactivated upon termination of the normal tissue repair
and clearance by immune cells via not yet fully understood mechanisms. in vivo, a phenotype characterized by reduction in α-SMA levels, con-
A growing body of evidence has reported that the senescence- tractile activities and ECM synthesis.79,80 The mechanisms promoting
associated secretory phenotype (SASP) of senescent myofibroblasts myofibroblast deactivation remain poorly defined, although several
includes pro-inflammatory chemokines and cytokines that lead to the reports have demonstrated a major role for peroxisome proliferator-
recruitment and activation of cytotoxic immune cells. Thus, senescent activated receptor gamma (PPARγ) at promoting myofibroblast deacti-
myofibroblasts appear to be programmed to be eliminated by the vation by blocking the TGF-β pathway. In the liver, 50% of total
immune system. This concept has been demonstrated in multiple studies myofibroblasts get deactivated during the resolution of murine liver
confirming the role of immune cells in eliminating senescent myo- fibrosis via up-regulation of PPARγ, albeit these reprogrammed cells
fibroblasts during tissue repair, and that impaired immune clearance of acquire a phenotype distinct from quiescent hepatic stellate cells.10,76
49,58–60
senescent cells leads to overwhelming fibrosis. Several immune In the lungs, lineage-tracing studies show that myofibroblasts get simi-
cell types have shown the capacity to eliminate senescent cells in vitro larly deactivated by turning into lipofibroblast-like cells during lung
and in vivo, including innate immune cells such as natural killer (NK) cells, fibrosis resolution, a process associated with PPARγ up-regulation.81 In
61 59,62,63
macrophages and neutrophils, but also T cells. In the context of the heart, periostin-traced myofibroblasts get deactivated to resting
wound healing, NK cells expressing TRAIL have been involved in the res- fibroblasts upon injury resolution in myocardial infarction models, a
olution of the tissue repair programme by promoting apoptosis of senes- mechanism associated with re-gaining expression of the transcription
44,49
cent myofibroblasts and other cell types. Similarly, cytotoxic CD4+ factor Tcf21.77 Accordingly, overexpression of Tcf21 deactivates
T cells expressing pro-apoptotic FasL have been shown to eliminate hepatic myofibroblasts in vitro and protects mice from CCl4-induced
myofibroblasts in the context of lung injury.45,64 In addition, senescent liver fibrosis and diet-induced steatohepatitis.82 In the skin, lineage-
dermal myofibroblasts can be cleared out by CD8+ T cells and NK cells tracing studies have also demonstrated that dermal myofibroblasts get
65
ex vivo. Mechanistically, the acquisition of a senescent phenotype deactivated via reprogramming to adipocytes through activation of the
leads to up-regulation of stress ligands in myofibroblasts and other cell BMP-ZFP423 pathway upon termination of the wound healing pro-
types, which can be recognized by specific receptors expressed in cyto- gramme.78 Further studies have shown that the transcription factor
toxic lymphocytes.66 Stress signals upregulated in senescent myo- MyoD acts as a critical switch controlling myofibroblasts activation and
fibroblasts, but not healthy cells, selectively target them for apoptosis by deactivation in vivo by modulating the TGF-β pathway.83 Together,
67–69
immune cells. For instance, up-regulation of MIC-A/B and ULBPs in these studies elegantly demonstrate the capacity of wound myo-
senescent myofibroblasts have been shown to activate both NK cells fibroblasts to get deactivated in vivo via reprogramming into resting
and a subset of CD8+ T cells via the NKG2D receptor.44,49,65,70 NKG2D cells or converting to another cell type. The molecular mediators
activation leads to NK and CD8+ T activation and the release of cyto- controlling these processes remain poorly understood, although
toxic granules containing perforin and granzymes that induce soluble factors such as PGE2, BMP4, PDGFB, HRAS, EGR4 and FGF-1
myofibroblast apoptosis. Accordingly, both NKG2D- and perforin- have been shown to block the TGF-β pathway in cultured myo-
deficient mice show accumulation of senescent liver myofibroblasts and fibroblasts.80,84,85 In addition, matrix softening has been also reported
increased fibrosis in mouse models of liver injury.64,70 More recent to induce myofibroblast deactivation. Several studies have shown that
reports have shown that senescent myofibroblasts may escape immune stiffness-activated myofibroblasts get deactivated when plated on soft
clearance in pathological settings by co-expressing inhibitory ligands or matrices.86–89 Similarly, desplinting and releasing the wound edge has
decoy receptors that block immune cell function. For instance, up- been shown to induce myofibroblast deactivation in vivo.29,90 In addition
64
regulation of the decoy death receptor Dcr2 or the inhibitory molecule to these cell-intrinsic mechanisms, bystander cells including immune cells
HLA-E65 in senescent myofibroblasts have been associated with resis- and adipocytes have been shown to promote myofibroblast deactivation.
tance to immune cell killing. In addition, leukocytes have been involved For instance, myofibroblasts in aortic valve stenosis get deactivated after
MERKT ET AL. 5

transcatheter aortic valve replacement and such deactivation is triggered pathways. The pro-fibrotic cytokine TGF-β promotes an apoptosis-
by IL-1β and TNF-α released from M1 macrophages in post-surgery resistant phenotype in myofibroblasts by activating the pro-survival
serum.91 Similarly, adipocyte-conditioned medium significantly decreases FAK-PI3K-AKT and p38 MAPK pathways.21,25,93 In addition, TGF-β
the expression of α-SMA and ECM components in skin myofibroblasts blocks mitochondrial apoptosis by modulating levels of the BCL-2
through BMP4 mediated activation of PPARγ signalling.85 More recent family of proteins. For instance, TGF-β inhibits the pro-apoptotic sen-
studies have shown that activated myofibroblast can get directly rep- sitizer protein BAD, upregulates pro-survival proteins such as BCL-2
rogrammed into adipocytes via BMP signalling during wound healing, and BCL-XL and decreases BAX to BCL-2 ratio via cellular Abelson (c-
contributing to tissue repair and regeneration.78 Abl) signalling.27,94–97 In addition to blocking the intrinsic apoptotic
pathway, TGF-β has been also shown to inhibit FasL-mediated activa-
tion of the extrinsic apoptotic pathways in scleroderma myo-
4 | M Y O F I B RO B L A S T P E RS I S T E N C E L E A D S fibroblasts by blocking ceramide production.94,98 Restoration of Fas
TO TISS UE FIBROSI S signalling upon treatment with PGE2, a lipid mediator which is found
deficient in IPF, increases Fas receptor expression and sensitizes myo-
Human fibrotic diseases including idiopathic pulmonary fibrosis (IPF), fibroblasts to apoptosis by inhibiting AKT signalling.99,100
systemic sclerosis (SSc, scleroderma), liver cirrhosis, cardiac fibrosis In addition to TGF-β signalling, increased matrix stiffness in fibrotic
and chronic kidney disease (CKD) are associated with high morbidity conditions has been shown to block apoptotic signalling in myofibroblasts
and mortality.92 Persistent myofibroblast activation is a hallmark of via activation of integrin-mediated mechanotransduction pathways.6,17 In
these fibrotic diseases and associates with progressive tissue scarring fibrotic diseases, myofibroblasts promote progressive matrix stiffening due
6,41
and organ failure. The molecular pathways that promote to excessive deposition and crosslinking of ECM proteins, locking them-
myofibroblast persistence beyond the resolution phase of tissue repair selves in a mechanical positive feedback loop. This vicious cycle amplifies
remain poorly understood, although recent studies have identified fibrotic responses by enhancing myofibroblast activation and persis-
mechanisms that keep myofibroblasts in an activated state by promot- tence.101 We and others have shown that stiff matrices directly activate
ing evasion of apoptosis, cellular senescence as well as metabolic and FAK, a pro-survival mechanosensitive protein activated by integrin signal-
epigenetic reprogramming (Figure 2). ling.22,102 FAK mediates Rho/Rho-associated protein kinase (ROCK) acti-
vation and increases acto-myosin contractility, which leads to activation of
transcriptional coactivators yes-associated protein (YAP)/transcriptional
4.1 | Evasion of apoptosis: Addiction to TGF-β1 coactivator with PDZ-binding motif (TAZ) and myocardin-related tran-
and mechanical signalling scription factor-A (MRTF-A)/MRTF-B, widely recognized as major
mechanotransducers.103,104 We and others have shown that stiffness-
Persistent growth factor signalling has been shown to promote activated YAP/TAZ promotes up-regulation of BCL-XL in myofibroblasts
myofibroblast evasion of apoptosis by activating pro-survival and that FAK–YAP–TAZ–BCL–XL pathway is hyperactivated in dermal

F I G U R E 2 Targeting myofibroblast fate for the treatment of established fibrosis. Myofibroblast persistence is a hallmark of fibrotic diseases
characterized by progressive tissue scarring. The molecular mechanisms that promote myofibroblast persistence include pathological growth
factor signalling (e.g. TGF-β pathway), mechanical signalling induced by matrix stiffness, cellular senescence and the senescence-associated
secretory phenotype (SASP: TGF-β, IL6, IL8) as well as metabolic and epigenetic reprogramming. This knowledge has paved the way for new
therapeutic strategies aiming at reversing fibrosis by targeting myofibroblast fate and promoting myofibroblast apoptosis and immune clearance,
deactivation and reprogramming. These novel treatments include senolytics, gene therapy, cellular immunotherapy and CAR-T cells, and may not
only reverse fibrosis but also regenerate fibrotic tissues
6 MERKT ET AL.

myofibroblasts from patients with scleroderma.15,105 Similarly, the scleroderma.118,125 Moreover, hypermethylation and subsequent
mechanosensitive pathway FAK–ROCK–MRTF–BCL-2 promotes sur- silencing of the proapoptotic protein P14ARF promotes an apoptosis-
106
vival and persistence of IPF fibroblasts. In addition, mechanical sig- resistant phenotype in IPF fibroblasts.126 Noteworthy, selective meth-
nalling inhibits the extrinsic apoptotic pathways by blocking acid ylation of three CpG islands in ACTA2 gene (the encoding gene of
sphingomyelinase and reducing Fas expression in myofibroblasts.28,94,98 α-SMA) has been also shown to reduce α-SMA expression in myo-
fibroblasts.127 On the contrary, DNA hypomethylation and conse-
quent transcription up-regulation of pro-fibrotic genes including
4.2 | Metabolic reprogramming COLIVA1, TGFBR3, SMAD3, SMAD6 has been described in kidney
fibrosis.121 Acetylation and methylation of histone proteins have been
Myofibroblasts undergo metabolic reprogramming in order to sustain similarly associated with changes in chromatin structure and increased
the high-energy demand associated with their ECM hyper-synthetic gene transcription in myofibroblasts. For instance, hyperacetylation of
and contractile phenotype. A growing body of evidence suggests that histone H4 by P300 has been shown to be required for TGF-
metabolic reprogramming, characterized by increased aerobic glycoly- β-induced type I collagen expression in myofibroblasts.128 Similarly,
sis, mitochondrial oxygen consumption and ROS production, pro- histone 3 lysine 9 (H3K9) hypermethylation of the PPARGC1A gene
19,107–112
motes myofibroblast persistence and survival. This notion promoter suppresses expression of PGC1α via the CBX5/G9a path-
that myofibroblast bioenergetics are linked to their persistent activa- way and promotes sustained activation of IPF fibroblasts.129 More-
tion state has been demonstrated by studies showing that inhibition over, decreased histone acetylation and increased histone 3 lysine
of glycolysis in myofibroblasts leads to rapid down-regulation of 9 trimethylation (H3K9Me3) of the Fas promoter reduces Fas expres-
α-SMA. 19,113
In addition, metabolic reprogramming and sion and promotes resistance to Fas-mediated apoptosis.130 Accord-
NOX4-mediated ROS production in myofibroblasts have been associ- ingly, pharmacological inhibition of histone deacetylases with
ated with mitochondrial dysfunction characterized by fragmented suberoylanilide hydroxamic acid (SAHA) reverses established lung fibro-
mitochondria and impaired activation of mitochondrial apoptosis in sis by inducing myofibroblast apoptosis via BCL-XL down-regulation.131
19,107,111
these cells. Accordingly, pharmacological activation of the On the contrary, histone deacetylation by the histone deacetylase
energy-sensor with metformin promotes mitochondrial biogenesis, 4 (HDAC4) has been reported to suppress the activated myofibroblast
restores myofibroblast sensitivity to apoptosis and reverses phenotype by inhibiting α-SMA expression.132 miRs suppress gene
established lung fibrosis in mouse models.18 Moreover, metabolic expression via direct binding to 30 UTR of target. In myofibroblasts,
reprogramming associates with enhanced glutamine metabolism in down-regulation of miR-29a and miR-133a has been associated with
IPF-derived fibroblasts, leading to increased expression of the apopto- myofibroblast persistence and survival, and that increase in miR-29a
sis inhibitors XIAP and surviving.114 More recently, increased promotes myofibroblast apoptosis.133 On the contrary, up-regulation of
glutaminolysis and glycolysis have been linked to Ca2+-dependent miR-21 has been shown to promote myofibroblast survival by reducing
metabolic reprogramming and myofibroblast persistence via epige- levels of pro-apoptotic BAX.134 Moreover, up-regulation of miR-22 has
115
netic mechanisms. been shown to promote myofibroblast persistence and survival by driv-
ing cellular senescence.135

4.3 | Epigenetic mechanisms


4.4 | Cellular senescence
Epigenetic modifications including DNA methylation, histone modifi-
cation and regulation by non-coding RNA such as microRNAs (miRs), Myofibroblast senescence has been traditionally associated with the
have been recently associated with myofibroblast activation and per- resolution of the normal tissue repair programme and that elimination
sistence.111,116 Difference in DNA methylation has been described in of senescent myofibroblasts prevents the development of fibro-
healthy control fibroblasts compared to fibroblasts isolated from sis.52,57 However, a growing body of evidence suggests that impaired
patients with IPF, CKD and scleroderma.117–122 Mechanistically, TGF- clearance of senescent myofibroblasts shifts their pro-repair activities
β signalling induces expression of DNA methyltransferase 3A to pro-fibrotic via persistent secretion of pro-inflammatory and pro-
(DNMT3A) and DNMT1 in myofibroblasts, leading to increased DNA fibrotic mediators50,136–141 . In this regard, the SASP of pro-fibrotic
123
methylation and modulation of pro-fibrotic gene expression. In this senescent myofibroblast has been shown to contain pro-inflammatory
regard, DNA hypermethylation silences endogenous anti-fibrotic cytokines (IL-1α, IL-1β, IL-6 and IL-8), pro-fibrotic cytokines (TGF-β,
genes such as PPARγ, a mechanism controlled by methyl cap binding PDGF) and ECM proteins (fibronectin, collagens).50,142 Human fibrotic
protein 2 (MeCP2), a transcriptional repressor that binds to methyl- fibroblasts derived from patients with various fibrotic diseases display
ated DNA.124 Accordingly, MeCP2-deficient mice showed protection a senescent phenotype.143–146 Noteworthy, senescent myofibroblasts
from fibrosis in the lungs and liver in mouse models.124 Similarly, activate pro-survival mechanisms including up-regulation of BCL-XL,
hypermethylation of anti-fibrotic genes such as THY-1 and FLI1 leads which promotes their persistence within fibrotic tissues. In humans,
to down-regulation of these molecules and increased type I collagen senescent myofibroblasts from patients with IPF show elevation of
synthesis in fibrotic fibroblasts isolated from patients with IPF and p16 and p21.50,51,55,136 Mechanistically, acquisition of a senescent
MERKT ET AL. 7

phenotype appears to be regulated by the DNA damage response,55 of ABT-737 and ABT-263 (navitoclax, which blocks BCL-2, BCL-XL,
independently from TGF-β signalling. BCL-W), ABT-199 (venetoclax, a BCL-2 selective inhibitor) and sev-
eral BCL-XL and MCL-1 mono-selective inhibitors.14,151–154 We have
recently shown that ABT-263 reverses established dermal fibrosis in a
5 | TA RG ETING M YO FIBROB LAS T mouse model of scleroderma by targeting myofibroblast for
P E R S I S T E N CE F O R A NT I - F I B RO TI C TH E R A P Y apoptosis.17 Mechanistically, ABT-263 directly blocks the pro-survival
protein BCL-XL, releasing pro-apoptotic proteins such as BIM that
Organ fibrosis has been traditionally thought to be irreversible. How- ultimately triggers myofibroblast apoptosis. In vitro, ABT-263 induces
ever, fibrosis has been shown to regress in humans in response to apoptosis of fibrotic fibroblasts isolated from patients with sclero-
pharmacological treatment of the underlying pro-fibrotic process. For derma.17 In addition, ABT-263 has been recently identified as a potent
instance, liver fibrosis can regress after treatment of viral hepatitis, senolytic agent by targeting senescent cells for apoptosis and revers-
pancreas transplantation can ameliorate diabetic nephropathy, hypo- ing lung fibrosis induced by ionizing radiation in mice.155 These data
tensive drugs can treat hypertensive cardiac fibrosis, and lung fibrosis validate genetic and pharmacological studies showing that elimination
can resolve substantially in patients with acute respiratory syndrome. of senescent cells protects mice from lung and kidney fibrosis.51,156–
158
These examples have in common that defined fibrogenic injuries exist, In the heart, ABT-263 has been shown to eliminate senescent
and that haltering of these pro-fibrotic insults associates with fibrosis cardiomyocytes and improve cardiac function in mouse models of car-
regression. However, the exact aetiology of most human fibrotic dis- diac ischemia–reperfusion injury159 and angiotensin II-induced cardiac
ease remains unknown such as in IPF or SSc. In these fibrotic diseases, hypertrophy and fibrosis.160 Importantly, clearance of senescent cells
it is thought that the establishment of self-amplification pro-fibrotic improved myocardial remodelling and diastolic activity along with a
loops leads to progressive fibrotic responses independently from the higher rate of survival in the event of myocardial infarction.161 In the
initial insult. Up to date, only nintedanib and pirfenidone have been liver, the BCL-XL selective inhibitor A-1331852 treats biliary liver
approved for the treatment of IPF and scleroderma-associated inter- fibrosis in a mouse model of primary sclerosing cholangitis by
stitial lung disease.147–149 Although these drugs slow down the rate targeting senescent cholangiocytes and fibroblasts for apoptosis.162
of progression of fibrosis, they do not halt or reverse established More recently, the MCL-1 inhibitor S63845 induces apoptosis of pro-
fibrosis.150 The mode of action of these drugs remains poorly under- liferative ductular reactive cells, reducing hepatic fibrosis in a mouse
stood, but they likely inhibit mechanisms involved in myofibroblast model of cholestasis.163
formation and collagen synthesis rather than mechanisms promoting
fibrosis resolution. Recent insights into the mechanisms promoting
myofibroblast persistence and survival have paved the way for new 5.1.2 | TRAIL ligands activate the extrinsic pathway
therapeutic strategies aiming at reversing fibrosis by targeting of apoptosis
myofibroblast fate. These novel treatments include senolytics, gene
therapy, cellular immunotherapy and CAR-T cells, and may not only As described above, several mechanisms contribute to reduced sensi-
reverse fibrosis but also regenerate fibrotic tissues (Figure 2). tivity of myofibroblasts to FasL-induced apoptosis.164 In vitro, activa-
tion of the cell-surface death receptors (DRs) of the extrinsic pathway
of apoptosis has been shown to sensitize myofibroblasts to FasL-
5.1 | Targeting myofibroblast apoptosis induced apoptosis.165 Accordingly, selective agonism of DR4 and DR5
with the TRAIL-TLY012 induces myofibroblast apoptosis and reverses
Substantial evidence has demonstrated that activated (senescent) established skin fibrosis in a mouse model of scleroderma,166
myofibroblasts in fibrotic tissues are prone to undergo apoptosis due highlighting the therapeutic potential of targeting myofibroblasts for
to increased mitochondrial priming, as described above. This knowl- apoptosis via activation of the extrinsic apoptosis pathway. Moreover,
edge of apoptotic priming has led to the development of novel anti- PEGylated TRAIL ameliorates liver cirrhosis in rats by inducing apo-
fibrotic strategies aimed at blocking pro-survival mechanisms acti- ptosis of activated hepatic stellate cells.167
vated in senescent myofibroblasts, ultimately inducing activation of
programmed cell death.
5.2 | Cellular immunotherapy

5.1.1 | Senolytic drugs unleash the intrinsic 5.2.1 | Harnessing macrophage and T cell anti-
pathway of apoptosis fibrotic potential

The so-called BH3 mimetic drugs are small molecules that selectively Harnessing the immune system to treat fibrosis has recently emerged
antagonize anti-apoptotic BCL-2 proteins (BCL-2, BCL-XL, BCL-W, as a promising therapeutic strategy. Recent reports have demonstrated
MCL-1 and A1) by directly binding to specific shallow hydrophobic that CD47, a “don't-eat-me-signal,” is upregulated in scleroderma myo-
grooves.14 This knowledge has led to the discovery and development fibroblasts and prevents phagocytosis by macrophages.168 Accordingly,
8 MERKT ET AL.

pharmacological blockade of CD47 leads to myofibroblast clearance by 5.4 | Targeting myofibroblast reprogramming
macrophages and reversion of fibrosis in an adaptive transfer model.169
Cytotoxic T cells have been similarly involved in myofibroblast clear- Conversion of activated myofibroblasts into epithelial-like cells holds
ance by activating the extrinsic pathway of apoptosis. In vivo, potential for the treatment and regeneration of fibrotic tissues. For
myofibroblast persist in FasL-deficient mice subjected to lung fibrosis instance, fibrogenic injury in the liver leads to loss of hepatocytes
models, and that reconstitution with FasL+ cytotoxic cells promote and accumulation of activated myofibroblasts. In this regard, recent
45
myofibroblast apoptosis and fibrosis resolution in mice. studies have demonstrated that direct reprogramming of liver myo-
fibroblasts into hepatocytes reverses established liver fibrosis and
repopulates the hepatocyte pool in mice. In these studies, lentiviral-
5.2.2 | CAR-T cells mediated ectopic overexpression of the transcription factors
FOXA3, GATA4, HNF1A and HNF4A converts mouse myo-
CAR-T cells are T-lymphocytes engineered to express a chimeric fibroblasts into hepatocyte-like cells and attenuates liver fibrosis.184
receptor targeting a specific antigen, thereby triggering T-cell activa- Similarly, liver myofibroblast reprogramming into hepatocytes using
tion and killing of target cells.170 Recently, CAR-T cells designed to adeno-associated virus (AAV) vectors expressing six hepatic tran-
selectivity eliminate cardiac fibroblasts expressing fibroblast activation scription factors including Foxa1, Foxa2, Foxa3, Gata4, Hnf1a or
protein (FAP) have been shown to reduce cardiac fibrosis and restora- Hnf4a reverses liver fibrosis in vivo.185 This novel therapeutic
171
tion of cardiac function after injury in mice. Similarly, CAR-T cells approach may open new avenues for the treatment of chronic liver
selectively targeting senescent cells expressing the urokinase-type fibrosis using gene therapy.
plasminogen activator receptor (uPAR) has been shown to treat liver
fibrosis in mouse models.172
6 | CONC LU SIONS

5.3 | Targeting myofibroblast deactivation Spatiotemporal control of myofibroblast functions is regulated by


alternative fates during normal tissue repair and fibrosis.
Over the last 20 years, multiple studies have demonstrated the ability of Myofibroblast deactivation, apoptosis, senescence and repro-
several agents at treating established fibrosis in preclinical models by pro- gramming appear to limit pro-repair functions and prevent the
moting myofibroblast deactivation to a low activity state. FDA-approved development of fibrosis: Whether these alternative myofibroblast
anti-fibrotic drugs, pirfenidone and nintedanib, have been shown to fates represent specific or redundant fail-safe mechanisms for lim-
reduce ECM synthesis and myofibroblast numbers by antagonizing the iting myofibroblast functions remains to be investigated. Thus, fur-
TGF-β pathway.173,174 Accordingly, direct inhibition of TGF-β ligand, ther research is needed to understand the mechanisms and
TGF-β receptors or integrin-mediated TGF-β activation have been simi- signalling pathways that determine these specific myofibroblast
larly shown to attenuate established fibrosis in mouse models.175 More- fates and their functions during normal tissue repair. In this line,
over, targeting secondary pro-fibrotic mediators induced by TGF-β such recent studies using single-cell RNA sequencing have shown that
as ephrin-B2, CTGF or ET-1 reduces ECM synthesis in myo- heterogeneous myofibroblast subpopulations undergo dynamic
15,176,177
fibroblasts. Similarly, antagonizing TGF-β signalling in fibroblasts phenotypic changes during tissue repair and fibrosis, further
with PPARγ agonists reduces matrix deposition both in vitro and suggesting that myofibroblast fate plasticity is essential for our
in vivo.175 In addition, inhibitors of mechanotransduction pathways body to respond to multiple injury scenarios. Despite these
including FAK, ROCK, YAP/TAZ and MRTFs have been shown to treat advances, it remains unknown how these different fates are inter-
12,178
established fibrosis by reducing myofibroblast pro-fibrotic activities. connected as well as their temporal and local wiring. A growing
Targeting epigenetic modulators such as the histone methylase SET8 body of evidence suggests that persistent myofibroblast activities
with the UNC0379 inhibitor or the bromodomain and extra-terminal lead to fibrosis, a manifestation of a pro-fibrotic phenotype associ-
(BET) with JQ1 have been shown to deactivate myofibroblasts and treat ated with impaired or chronic myofibroblast fate. Therapeutic
179,180
pre-clinical fibrosis. The list of anti-fibrotic targets aimed at reduc- targeting of myofibroblast fates such as induction of apoptosis of
ing myofibroblast activities is evergrowing and extensible reviewed else- (senescent) myofibroblasts, deactivation or reprograming could
where. More recent studies have identified functionally and spatially reverse established fibrosis and potentially regenerate chronically
distinct fibroblast lineages involved in skin repair and fibrosis, supporting injured tissues. Given that the origin of myofibroblasts depends on
the notion that specific pro-fibrotic fibroblasts populations can be specific progenitors present in each tissue (e.g. resident fibroblasts,
targeted for therapeutic treatment.181–183 Specifically, the Engrailed-1 epithelial and endothelial cells, pericytes and adipocytes), it will be
fibroblast lineage is endowed with fibrogenic potential and is the primary important to understand the contribution and function of specific
contributor to ECM synthesis during embryonic development, wounding myofibroblast fates in different organs and systems, which might
181
and fibrosis. Accordingly, selective targeting of Engrailed-1+ fibro- inform about organ-specific anti-fibrotic strategies. Further
blasts with the CD26 pharmacological inhibitor diprotin A reduced skin research is needed to translate these innovative and promising
scarring in wounding models.181 therapeutic strategies from the bench to the bedside.
MERKT ET AL. 9

ACKNOWLEDGMENTS 13. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol.
D.L. gratefully acknowledges funding support from the NIH (grant 2008;214:199-210. https://doi.org/10.1002/path.2277
14. Kuehl T, Lagares D. BH3 mimetics as anti-fibrotic therapy:
R01 HL147059-01) and Sponsored Research Grants from Boehringer
unleashing the mitochondrial pathway of apoptosis in myo-
Ingelheim and Merck & Co. fibroblasts. Matrix Biol. 2018;68-69:94-105. https://doi.org/10.
1016/j.matbio.2018.01.020
CONF LICT OF IN TR ES T 15. Lagares D, Ghassemi-Kakroodi P, Tremblay C, et al.
ADAM10-mediated ephrin-B2 shedding promotes myofibroblast
D.L. declares that he has received research funding from Boehringer
activation and organ fibrosis. Nat Med. 2017;23:1405-1415. https://
Ingelheim, Indalo Therapeutics, Merck & Co and Unity Biotechnology. doi.org/10.1038/nm.4419
Dr. Lagares has a financial interest in Mediar Therapeutics and Zenon 16. Youle RJ, Strasser A. The BCL-2 protein family: opposing activities
Biotech. The companies are developing treatments for organ fibrosis. that mediate cell death. Nat Rev Mol Cell Biol. 2008;9:47-59. https://
doi.org/10.1038/nrm2308
Dr. Lagares's interests were reviewed and are managed by MGH and
17. Lagares D, Santos A, Grasberger PE, et al. Targeted apoptosis of
Partners HealthCare in accordance with their conflict of interest poli- myofibroblasts with the BH3 mimetic ABT-263 reverses established
cies. WM, YZ and HH declare no conflict of interest. fibrosis. Sci Transl Med. 2017;9:eaal3765. https://doi.org/10.1126/
scitranslmed.aal3765
18. Rangarajan S, Bone NB, Zmijewska AA, et al. Metformin reverses
DATA AVAI LAB ILITY S TATEMENT
established lung fibrosis in a bleomycin model. Nat Med. 2018;24:
The data that support the concepts described in this manuscript is
1121-1127. https://doi.org/10.1038/s41591-018-0087-6
derived from resources available in the public domain. 19. Bernard K, Logsdon NJ, Ravi S, et al. Metabolic reprogramming is
required for myofibroblast contractility and differentiation. J Biol
ORCID Chem. 2015;290:25427-25438. https://doi.org/10.1074/jbc.M115.
646984
David Lagares https://orcid.org/0000-0002-7317-8796
20. Rizvi S, Mertens JC, Bronk SF, et al. Platelet-derived growth
factor primes cancer-associated fibroblasts for apoptosis. J Biol
RE FE R ENC E S Chem. 2014;289:22835-22849. https://doi.org/10.1074/jbc.M114.
1. Darby IA, Laverdet B, Bonte F, Desmouliere A. Fibroblasts and myo- 563064
fibroblasts in wound healing. Clin Cosmet Investig Dermatol. 2014;7: 21. Horowitz JC, Rogers DS, Sharma V, et al. Combinatorial activation of
301-311. https://doi.org/10.2147/CCID.S50046 FAK and AKT by transforming growth factor-beta1 confers an
anoikis-resistant phenotype to myofibroblasts. Cell Signal. 2007;19:
2. Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myo-
761-771. https://doi.org/10.1016/j.cellsig.2006.10.001
fibroblasts and mechano-regulation of connective tissue remodelling. Nat
22. Lagares D, Busnadiego O, García-Fernandez RA, et al. Inhibition of
Rev Mol Cell Biol. 2002;3:349-363. https://doi.org/10.1038/nrm809
focal adhesion kinase prevents experimental lung fibrosis and
3. Rockey DC, Bell PD, Hill JA. Fibrosis—a common pathway to organ
myofibroblast formation. Arthritis Rheum. 2012;64:1653-1664.
injury and failure. N Engl J Med. 2015;373:95-96. https://doi.org/10.
https://doi.org/10.1002/art.33482
1056/NEJMc1504848
23. Horowitz JC, Ajayi IO, Kulasekaran P, et al. Survivin expression
4. Darby I, Skalli O, Gabbiani G. Alpha-smooth muscle actin is tran-
induced by endothelin-1 promotes myofibroblast resistance to apo-
siently expressed by myofibroblasts during experimental wound
ptosis. Int J Biochem Cell Biol. 2012;44:158-169. https://doi.org/10.
healing. Lab Invest. 1990;63:21-29.
1016/j.biocel.2011.10.011
5. Iredale JP, Benyon RC, Pickering J, et al. Mechanisms of spontane-
24. Kulkarni AA, Thatcher TH, Olsen KC, Maggirwar SB, Phipps RP,
ous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis and
Sime PJ. PPAR-gamma ligands repress TGFbeta-induced myofibroblast
reduced hepatic expression of metalloproteinase inhibitors. J Clin
differentiation by targeting the PI3K/Akt pathway: implications for
Invest. 1998;102:538-549. https://doi.org/10.1172/JCI1018
therapy of fibrosis. PLoS One. 2011;6:e15909. https://doi.org/10.
6. Hinz B, Lagares D. Evasion of apoptosis by myofibroblasts: a hall-
1371/journal.pone.0015909
mark of fibrotic diseases. Nat Rev Rheumatol. 2020;16:11-31.
25. Kulasekaran P, Scavone CA, Rogers DS, Arenberg DA,
https://doi.org/10.1038/s41584-019-0324-5
Thannickal VJ, Horowitz JC. Endothelin-1 and transforming growth
7. Desmouliere A, Redard M, Darby I, Gabbiani G. Apoptosis mediates
factor-beta1 independently induce fibroblast resistance to apoptosis
the decrease in cellularity during the transition between granulation
via AKT activation. Am J Respir Cell Mol Biol. 2009;41:484-493.
tissue and scar. Am J Pathol. 1995;146:56-66.
https://doi.org/10.1165/rcmb.2008-0447OC
8. Redente EF, Chakraborty S, Sajuthi S et al. Loss of Fas signaling in
26. Lagares D, Busnadiego O, Garcia-Fernandez RA, Lamas S,
fibroblasts impairs homeostatic fibrosis resolution and promotes per-
Rodriguez-Pascual F. Adenoviral gene transfer of endothelin-1 in the
sistent pulmonary fibrosis. JCI Insight. 2020;6:e141618. https://doi.
lung induces pulmonary fibrosis through the activation of focal
org/10.1172/jci.insight.141618
adhesion kinase. Am J Respir Cell Mol Biol. 2012;47:834-842.
9. Liu X, Wong SS, Taype CA, et al. Thy-1 interaction with Fas in lipid rafts
https://doi.org/10.1165/rcmb.2011-0446OC
regulates fibroblast apoptosis and lung injury resolution. Lab Invest.
27. Zhang HY, Phan SH. Inhibition of myofibroblast apoptosis by trans-
2017;97:256-267. https://doi.org/10.1038/labinvest.2016.145
forming growth factor beta(1). Am J Respir Cell Mol Biol. 1999;21:
10. Kisseleva T, Cong M, Paik Y, et al. Myofibroblasts revert to an inac-
658-665. https://doi.org/10.1165/ajrcmb.21.6.3720
tive phenotype during regression of liver fibrosis. Proc Natl Acad Sci.
28. Samuel GH, Lenna S, Bujor AM, Lafyatis R, Trojanowska M. Acid
2012;109:9448-9453. https://doi.org/10.1073/pnas.1201840109
sphingomyelinase deficiency contributes to resistance of sclero-
11. Hinz B. Myofibroblasts. Exp Eye Res. 2016;142:56-70. https://doi.
derma fibroblasts to Fas-mediated apoptosis. J Dermatol Sci.
org/10.1016/j.exer.2015.07.009
2012;67:166-172. https://doi.org/10.1016/j.jdermsci.2012.
12. Santos A, Lagares D. Matrix stiffness: the conductor of organ fibrosis.
06.001
Curr Rheumatol Rep. 2018;20:2. https://doi.org/10.1007/s11926-
29. Hinz B, Mastrangelo D, Iselin CE, Chaponnier C, Gabbiani G.
018-0710-z
Mechanical tension controls granulation tissue contractile activity
10 MERKT ET AL.

and myofibroblast differentiation. Am J Pathol. 2001;159:1009- 46. Polunovsky VA, Chen B, Henke C, et al. Role of mesenchymal cell
1020. death in lung remodeling after injury. J Clin Invest. 1993;92:388-397.
30. Van De Water L, Varney S, Tomasek JJ. Mechanoregulation of the https://doi.org/10.1172/JCI116578
myofibroblast in wound contraction, scarring, and fibrosis: opportu- 47. Davan-Wetton CSA, Pessolano E, Perretti M, Montero-Melendez T.
nities for new therapeutic intervention. Adv Wound Care (New Senescence under appraisal: hopes and challenges revisited. Cell Mol Life
Rochelle). 2013;2:122-141. https://doi.org/10.1089/wound.2012. Sci. 2021;78:3333-3354. https://doi.org/10.1007/s00018-020-03746-x
0393 48. Munoz-Espin D, Serrano M. Cellular senescence: from physiology to
31. Grinnell F, Zhu M, Carlson MA, Abrams JM. Release of mechanical pathology. Nat Rev Mol Cell Biol. 2014;15:482-496. https://doi.org/
tension triggers apoptosis of human fibroblasts in a model of 10.1038/nrm3823
regressing granulation tissue. Exp Cell Res. 1999;248:608-619. 49. Krizhanovsky V, Yon M, Dickins RA, et al. Senescence of activated
https://doi.org/10.1006/excr.1999.4440 stellate cells limits liver fibrosis. Cell. 2008;134:657-667. https://doi.
32. Linge C, Richardson J, Vigor C, Clayton E, Hardas B, Rolfe KJ. Hyper- org/10.1016/j.cell.2008.06.049
trophic scar cells fail to undergo a form of apoptosis specific to con- 50. Alvarez D, Cardenes N, Sellarés J, et al. IPF lung fibroblasts have a
tractile collagen—the role of tissue transglutaminase. J Invest senescent phenotype. Am J Physiol Lung Cell Mol Physiol. 2017;313:
Dermatol. 2005;125:72-82. https://doi.org/10.1111/j.0022-202X. L1164-L1173. https://doi.org/10.1152/ajplung.00220.2017
2005.23771.x 51. Schafer MJ, White TA, Iijima K, et al. Cellular senescence mediates
33. Kural MH, Billiar KL. Myofibroblast persistence with real-time fibrotic pulmonary disease. Nat Commun. 2017;8:14532. https://doi.
changes in boundary stiffness. Acta Biomater. 2016;32:223-230. org/10.1038/ncomms14532
https://doi.org/10.1016/j.actbio.2015.12.031 52. Jun JI, Lau LF. The matricellular protein CCN1 induces fibroblast
34. Carlson MA, Longaker MT, Thompson JS. Wound splinting regulates senescence and restricts fibrosis in cutaneous wound healing. Nat
granulation tissue survival. J Surg Res. 2003;110:304-309. https:// Cell Biol. 2010;12:676-685. https://doi.org/10.1038/ncb2070
doi.org/10.1016/s0022-4804(02)00098-7 53. Demaria M, Ohtani N, Youssef SA, et al. An essential role for
35. Qu J, Yang S-Z, Zhu Y, Guo T, Thannickal VJ, Zhou Y. Targeting senescent cells in optimal wound healing through secretion of
mechanosensitive MDM4 promotes lung fibrosis resolution in aged PDGF-AA. Dev Cell. 2014;31:722-733. https://doi.org/10.1016/j.
mice. J Exp Med. 2021;218:e20202033. https://doi.org/10.1084/ devcel.2014.11.012
jem.20202033 54. Wolstein JM, Lee DH, Michaud J, Buot V, Stefanchik B, Plotkin MD.
36. Duffield JS, Forbes SJ, Constandinou CM, et al. Selective depletion INK4a knockout mice exhibit increased fibrosis under normal condi-
of macrophages reveals distinct, opposing roles during liver injury tions and in response to unilateral ureteral obstruction. Am J Physiol
and repair. J Clin Invest. 2005;115:56-65. https://doi.org/10.1172/ Renal Physiol. 2010;299:F1486-F1495. https://doi.org/10.1152/
JCI22675 ajprenal.00378.2010
37. Ramachandran P, Pellicoro A, Vernon MA, et al. Differential Ly-6C 55. Pitiyage GN, Slijepcevic P, Gabrani A, et al. Senescent mesenchymal
expression identifies the recruited macrophage phenotype, which cells accumulate in human fibrosis by a telomere-independent mech-
orchestrates the regression of murine liver fibrosis. Proc Natl Acad anism and ameliorate fibrosis through matrix metalloproteinases.
Sci U S A. 2012;109:E3186-E3195. https://doi.org/10.1073/pnas. J Pathol. 2011;223:604-617. https://doi.org/10.1002/path.2839
1119964109 56. Fitzner B, Müller S, Walther M, et al. Senescence determines the
38. Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, et al. Identification of a fate of activated rat pancreatic stellate cells. J Cell Mol Med.
novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti- 2012;16:2620-2630. https://doi.org/10.1111/j.1582-4934.
apoptotic factors. Aging Cell. 2016;15:428-435. https://doi.org/10. 2012.01573.x
1111/acel.12445 57. Kim K-H, Chen C-C, Monzon RI, Lau LF. Matricellular protein CCN1
39. Bundesmann MM, Wagner TE, Chow YH, Altemeier WA, promotes regression of liver fibrosis through induction of cellular
Steinbach T, Schnapp LM. Role of urokinase plasminogen activator senescence in hepatic myofibroblasts. Mol Cell Biol. 2013;33:2078-
receptor-associated protein in mouse lung. Am J Respir Cell Mol Biol. 2090. https://doi.org/10.1128/MCB.00049-13
2012;46:233-239. https://doi.org/10.1165/rcmb.2010-0485OC 58. Sagiv A, Krizhanovsky V. Immunosurveillance of senescent cells: the
40. Atabai K, Jame S, Azhar N, et al. Mfge8 diminishes the severity of bright side of the senescence program. Biogerontology. 2013;14:
tissue fibrosis in mice by binding and targeting collagen for uptake 617-628. https://doi.org/10.1007/s10522-013-9473-0
by macrophages. J Clin Invest. 2009;119:3713-3722. https://doi. 59. Song P, An J, Zou MH. Immune clearance of senescent cells to com-
org/10.1172/JCI40053 bat ageing and chronic diseases. Cells. 2020;9:671. https://doi.org/
41. Horowitz JC, Thannickal VJ. Mechanisms for the resolution of organ 10.3390/cells9030671
fibrosis. Physiology (Bethesda). 2019;34:43-55. https://doi.org/10. 60. Binet F, Cagnone G, Crespo-Garcia S, et al. Neutrophil extracellular
1152/physiol.00033.2018 traps target senescent vasculature for tissue remodeling in retinopa-
42. Frangogiannis N. Transforming growth factor-beta in tissue fibrosis. thy. Science. 2020;369:eaay5356. https://doi.org/10.1126/science.
J Exp Med. 2020;217:e20190103. https://doi.org/10.1084/jem. aay5356
20190103 61. Xue W, Zender L, Miething C, et al. Senescence and tumour clear-
43. Kaur H, Chaurasia SS, Agrawal V, Suto C, Wilson SE. Corneal ance is triggered by p53 restoration in murine liver carcinomas.
myofibroblast viability: opposing effects of IL-1 and TGF beta1. Exp Nature. 2007;445:656-660. https://doi.org/10.1038/nature05529
Eye Res. 2009;89:152-158. https://doi.org/10.1016/j.exer.2009. 62. Antonangeli F, Zingoni A, Soriani A, Santoni A. Senescent cells: living
03.001 or dying is a matter of NK cells. J Leukoc Biol. 2019;105:1275-1283.
44. Radaeva S, Sun R, Jaruga B, Nguyen VT, Tian Z, Gao B. Natural killer https://doi.org/10.1002/JLB.MR0718-299R
cells ameliorate liver fibrosis by killing activated stellate cells in 63. Hoenicke L, Zender L. Immune surveillance of senescent cells—
NKG2D-dependent and tumor necrosis factor-related apoptosis- biological significance in cancer- and non-cancer pathologies. Carci-
inducing ligand-dependent manners. Gastroenterology. 2006;130: nogenesis. 2012;33:1123-1126. https://doi.org/10.1093/carcin/
435-452. https://doi.org/10.1053/j.gastro.2005.10.055 bgs124
45. Wallach-Dayan SB, Elkayam L, Golan-Gerstl R, et al. Cutting edge: 64. Sagiv A, Biran A, Yon M, Simon J, Lowe SW, Krizhanovsky V. Granule
FasL(+) immune cells promote resolution of fibrosis. J Autoimmun. exocytosis mediates immune surveillance of senescent cells. Oncogene.
2015;59:67-76. https://doi.org/10.1016/j.jaut.2015.02.006 2013;32:1971-1977. https://doi.org/10.1038/onc.2012.206
MERKT ET AL. 11

65. Pereira BI, Devine OP, Vukmanovic-Stejic M, et al. Senescent cells 83. Hecker L, Jagirdar R, Jin T, Thannickal VJ. Reversible differentiation
evade immune clearance via HLA-E-mediated NK and CD8(+) T cell of myofibroblasts by MyoD. Exp Cell Res. 2011;317:1914-1921.
inhibition. Nat Commun. 2019;10:2387. https://doi.org/10.1038/ https://doi.org/10.1016/j.yexcr.2011.03.016
s41467-019-10335-5 84. Kosla J, Dvorakova M, Dvorak M, Cermak V. Effective myofibroblast
66. Soriani A, Zingoni A, Cerboni C, et al. ATM-ATR-dependent up- dedifferentiation by concomitant inhibition of TGF-β signaling and
regulation of DNAM-1 and NKG2D ligands on multiple myeloma perturbation of MAPK signaling. Eur J Cell Biol. 2013;92:363-373.
cells by therapeutic agents results in enhanced NK-cell suscepti- https://doi.org/10.1016/j.ejcb.2013.10.013
bility and is associated with a senescent phenotype. Blood. 2009; 85. Hoerst K, van den Broek L, Sachse C, et al. Regenerative potential of
113:3503-3511. https://doi.org/10.1182/blood-2008-08- adipocytes in hypertrophic scars is mediated by myofibroblast repro-
173914 gramming. J Mol Med (Berl). 2019;97:761-775. https://doi.org/10.
67. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of 1007/s00109-019-01772-2
natural killer cells. Nat Immunol. 2008;9:503-510. https://doi.org/ 86. Liu F, Lagares D, Choi KM, et al. Mechanosignaling through YAP and
10.1038/ni1582 TAZ drives fibroblast activation and fibrosis. Am J Physiol Lung Cell
68. Vivier E, Raulet DH, Moretta A, et al. Innate or adaptive immunity? Mol Physiol. 2015;308:L344-L357. https://doi.org/10.1152/ajplung.
The example of natural killer cells. Science. 2011;331:44-49. https:// 00300.2014
doi.org/10.1126/science.1198687 87. Tschumperlin DJ, Liu F, Tager AM. Biomechanical regulation of mes-
69. Chan CJ, Smyth MJ, Martinet L. Molecular mechanisms of natural enchymal cell function. Curr Opin Rheumatol. 2013;25:92-100.
killer cell activation in response to cellular stress. Cell Death Differ. https://doi.org/10.1097/BOR.0b013e32835b13cd
2014;21:5-14. https://doi.org/10.1038/cdd.2013.26 88. Wang H, Haeger SM, Kloxin AM, Leinwand LA, Anseth KS. Red-
70. Sagiv A, Burton DG, Moshayev Z, et al. NKG2D ligands mediate irecting valvular myofibroblasts into dormant fibroblasts through
immunosurveillance of senescent cells. Aging (Albany NY). 2016;8: light-mediated reduction in substrate modulus. PLoS One. 2012;7:
328-344. https://doi.org/10.18632/aging.100897 e39969. https://doi.org/10.1371/journal.pone.0039969
71. Jeong WI, Park O, Gao B. Abrogation of the antifibrotic effects of 89. Granato G, Ruocco MR, Iaccarino A, et al. Generation and analysis of
natural killer cells/interferon-gamma contributes to alcohol accelera- spheroids from human primary skin myofibroblasts: an experimental
tion of liver fibrosis. Gastroenterology. 2008;134:248-258. https:// system to study myofibroblasts deactivation. Cell Death Discov.
doi.org/10.1053/j.gastro.2007.09.034 2017;3:17038. https://doi.org/10.1038/cddiscovery.2017.38
72. Jeong WI, Park O, Suh YG, et al. Suppression of innate immunity 90. Wipff PJ, Rifkin DB, Meister JJ, Hinz B. Myofibroblast contraction
(natural killer cell/interferon-gamma) in the advanced stages of liver activates latent TGF-beta1 from the extracellular matrix. J Cell Biol.
fibrosis in mice. Hepatology. 2011;53:1342-1351. https://doi.org/ 2007;179:1311-1323. https://doi.org/10.1083/jcb.200704042
10.1002/hep.24190 91. Aguado BA, Schuetze KB, Grim JC, et al. Transcatheter aortic valve
73. Bansal R, Prakash J, Post E, Beljaars L, Schuppan D, Poelstra K. replacements alter circulating serum factors to mediate
Novel engineered targeted interferon-gamma blocks hepatic myofibroblast deactivation. Sci Transl Med. 2019;11:eaav3233.
fibrogenesis in mice. Hepatology. 2011;54:586-596. https://doi.org/ https://doi.org/10.1126/scitranslmed.aav3233
10.1002/hep.24395 92. Rockey DC, Bell PD, Hill JA. Fibrosis—a common pathway to organ
74. Nagata S. Apoptosis and clearance of apoptotic cells. Annu Rev Immunol. injury and failure. N Engl J Med. 2015;372:1138-1149. https://doi.
2018;36:489-517. https://doi.org/10.1146/annurev-immunol-042617- org/10.1056/NEJMra1300575
053010 93. Horowitz JC, Lee DY, Waghray M, et al. Activation of the pro-
75. Doran AC, Yurdagul A Jr, Tabas I. Efferocytosis in health and dis- survival phosphatidylinositol 3-kinase/AKT pathway by transforming
ease. Nat Rev Immunol. 2020;20:254-267. https://doi.org/10.1038/ growth factor-beta1 in mesenchymal cells is mediated by p38
s41577-019-0240-6 MAPK-dependent induction of an autocrine growth factor. J Biol
76. Troeger JS, Mederacke I, Gwak GY, et al. Deactivation of hepatic Chem. 2004;279:1359-1367. https://doi.org/10.1074/jbc.
stellate cells during liver fibrosis resolution in mice. Gastroenterology. M306248200
2012;143:1073-1083.e22. https://doi.org/10.1053/j.gastro.2012. 94. Jelaska A, Korn JH. Role of apoptosis and transforming growth fac-
06.036 tor beta1 in fibroblast selection and activation in systemic sclerosis.
77. Kanisicak O, Khalil H, Ivey MJ, et al. Genetic lineage tracing defines Arthritis Rheum. 2000;43:2230-2239. https://doi.org/10.1002/
myofibroblast origin and function in the injured heart. Nat Commun. 1529-0131(200010)43:10<2230::Aid-anr10>3.0.Co;2-8
2016;7:12260. https://doi.org/10.1038/ncomms12260 95. Zhang L, Zhou F, ten Dijke P. Signaling interplay between trans-
78. Plikus MV, Guerrero-Juarez CF, Ito M, et al. Regeneration of fat cells forming growth factor-β receptor and PI3K/AKT pathways in cancer.
from myofibroblasts during wound healing. Science. 2017;355:748- Trends Biochem Sci. 2013;38:612-620. https://doi.org/10.1016/j.
752. https://doi.org/10.1126/science.aai8792 tibs.2013.10.001
79. Gerarduzzi C, Di Battista JA. Myofibroblast repair mechanisms post- 96. Karimizadeh E, Gharibdoost F, Motamed N, Jafarinejad-Farsangi S,
inflammatory response: a fibrotic perspective. Inflamm Res. 2017;66: Jamshidi A, Mahmoudi M. c-Abl silencing reduced the inhibitory
451-465. https://doi.org/10.1007/s00011-016-1019-x effects of TGF-β1 on apoptosis in systemic sclerosis dermal fibro-
80. Wettlaufer SH, Scott JP, McEachin RC, Peters-Golden M, Huang SK. blasts. Mol Cell Biochem. 2015;405:169-176. https://doi.org/10.
Reversal of the transcriptome by prostaglandin E2 during 1007/s11010-015-2408-0
myofibroblast dedifferentiation. Am J Respir Cell Mol Biol. 2016;54: 97. Boström H, Willetts K, Pekny M, et al. PDGF-A signaling is a critical
114-127. https://doi.org/10.1165/rcmb.2014-0468OC event in lung alveolar myofibroblast development and alveogenesis.
81. El Agha E, Moiseenko A, Kheirollahi V, et al. Two-way conversion Cell. 1996;85:863-873. https://doi.org/10.1016/s0092-8674(00)
between lipogenic and myogenic fibroblastic phenotypes marks the 81270-2
progression and resolution of lung fibrosis. Cell Stem Cell. 2017;20: 98. Santiago B, Galindo M, Rivero M, Pablos JL. Decreased susceptibility
261-273.e263. https://doi.org/10.1016/j.stem.2016.10.004 to Fas-induced apoptosis of systemic sclerosis dermal fibroblasts.
82. Nakano Y, Kamiya A, Sumiyoshi H, Tsuruya K, Kagawa T, Inagaki Y. Arthritis Rheum. 2001;44:1667-1676. https://doi.org/10.1002/
A deactivation factor of fibrogenic hepatic stellate cells induces 1529-0131(200107)44:7<1667::Aid-art291>3.0.Co;2-y
regression of liver fibrosis in mice. Hepatology. 2020;71:1437-1452. 99. Huang S, Wettlaufer SH, Hogaboam C, Aronoff DM, Peters-
https://doi.org/10.1002/hep.30965 Golden M. Prostaglandin E(2) inhibits collagen expression and
12 MERKT ET AL.

proliferation in patient-derived normal lung fibroblasts via E 116. Duong TE, Hagood JS. Epigenetic regulation of myofibroblast phe-
prostanoid 2 receptor and cAMP signaling. Am J Physiol Lung Cell notypes in fibrosis. Curr Pathobiol Rep. 2018;6:79-96. https://doi.
Mol Physiol. 2007;292:L405-L413. https://doi.org/10.1152/ajplung. org/10.1007/s40139-018-0155-0
00232.2006 117. Huang SK, Scruggs AM, McEachin RC, White ES, Peters-Golden M.
100. Maher TM, Evans IC, Bottoms SE, et al. Diminished prostaglandin E2 Lung fibroblasts from patients with idiopathic pulmonary fibrosis
contributes to the apoptosis paradox in idiopathic pulmonary fibro- exhibit genome-wide differences in DNA methylation compared to
sis. Am J Respir Crit Care Med. 2010;182:73-82. https://doi.org/10. fibroblasts from nonfibrotic lung. PLoS One. 2014;9:e107055.
1164/rccm.200905-0674OC https://doi.org/10.1371/journal.pone.0107055
101. Liu F, Mih JD, Shea BS, et al. Feedback amplification of fibrosis 118. Sanders YY, Ambalavanan N, Halloran B, et al. Altered DNA methyl-
through matrix stiffening and COX-2 suppression. J Cell Biol. 2010; ation profile in idiopathic pulmonary fibrosis. Am J Respir Crit Care
190:693-706. https://doi.org/10.1083/jcb.201004082 Med. 2012;186:525-535. https://doi.org/10.1164/rccm.201201-
102. Lagares D, Kapoor M. Targeting focal adhesion kinase in fibrotic dis- 0077OC
eases. BioDrugs. 2013;27:15-23. https://doi.org/10.1007/s40259- 119. Rabinovich EI, Kapetanaki MG, Steinfeld I, et al. Global methylation
012-0003-4 patterns in idiopathic pulmonary fibrosis. PLoS One. 2012;7:e33770.
103. Dupont S, Morsut L, Aragona M, et al. Role of YAP/TAZ in https://doi.org/10.1371/journal.pone.0033770
mechanotransduction. Nature. 2011;474:179-183. https://doi.org/ 120. Yang IV, Pedersen BS, Rabinovich E, et al. Relationship of DNA
10.1038/nature10137 methylation and gene expression in idiopathic pulmonary fibrosis.
104. Xu H, Wu X, Qin H, et al. Myocardin-related transcription factor A Am J Respir Crit Care Med. 2014;190:1263-1272. https://doi.org/10.
epigenetically regulates renal fibrosis in diabetic nephropathy. J Am 1164/rccm.201408-1452OC
Soc Nephrol. 2015;26:1648-1660. https://doi.org/10.1681/ASN. 121. Ko YA, Mohtat D, Suzuki M, et al. Cytosine methylation changes in
2014070678 enhancer regions of core pro-fibrotic genes characterize kidney
105. Seo J, Kim J. Regulation of Hippo signaling by actin remodeling. fibrosis development. Genome Biol. 2013;14:R108. https://doi.org/
BMB Rep. 2018;51:151-156. https://doi.org/10.5483/bmbrep.2018. 10.1186/gb-2013-14-10-r108
51.3.012 122. Altorok N, Tsou PS, Coit P, Khanna D, Sawalha AH. Genome-wide
106. Zhou Y, Huang X, Hecker L, et al. Inhibition of mechanosensitive sig- DNA methylation analysis in dermal fibroblasts from patients with
naling in myofibroblasts ameliorates experimental pulmonary fibro- diffuse and limited systemic sclerosis reveals common and subset-
sis. J Clin Invest. 2013;123:1096-1108. https://doi.org/10.1172/ specific DNA methylation aberrancies. Ann Rheum Dis. 2015;74:
jci66700 1612-1620. https://doi.org/10.1136/annrheumdis-2014-205303
107. Xie N, Tan Z, Banerjee S, et al. Glycolytic reprogramming in 123. Koh HB, Scruggs AM, Huang SK. Transforming growth factor-beta1
myofibroblast differentiation and lung fibrosis. Am J Respir Crit Care increases DNA methyltransferase 1 and 3a expression through dis-
Med. 2015;192:1462-1474. https://doi.org/10.1164/rccm.201504- tinct post-transcriptional mechanisms in lung fibroblasts. J Biol
0780OC Chem. 2016;291:19287-19298. https://doi.org/10.1074/jbc.M116.
108. Negmadjanov U, Godic Z, Rizvi F, et al. TGF-β1-mediated differenti- 723080
ation of fibroblasts is associated with increased mitochondrial con- 124. Mann J, Chu DC, Maxwell A, et al. MeCP2 controls an epigenetic
tent and cellular respiration. PLoS One. 2015;10:e0123046. https:// pathway that promotes myofibroblast transdifferentiation and fibro-
doi.org/10.1371/journal.pone.0123046 sis. Gastroenterology. 2010;138:705-714, 714.e701-704. https://
109. Sampson N, Koziel R, Zenzmaier C, et al. ROS signaling by NOX4 doi.org/10.1053/j.gastro.2009.10.002
drives fibroblast-to-myofibroblast differentiation in the diseased 125. Wang Y, Fan PS, Kahaleh B. Association between enhanced type I
prostatic stroma. Mol Endocrinol. 2011;25:503-515. https://doi.org/ collagen expression and epigenetic repression of the FLI1 gene in
10.1210/me.2010-0340 scleroderma fibroblasts. Arthritis Rheum. 2006;54:2271-2279.
110. Park IH, Park SJ, Cho JS, et al. Role of reactive oxygen species in https://doi.org/10.1002/art.21948
transforming growth factor beta1-induced alpha smooth-muscle 126. Cisneros J, Hagood J, Checa M, et al. Hypermethylation-mediated
actin and collagen production in nasal polyp-derived fibroblasts. Int silencing of p14(ARF) in fibroblasts from idiopathic pulmonary fibro-
Arch Allergy Immunol. 2012;159:278-286. https://doi.org/10.1159/ sis. Am J Physiol Lung Cell Mol Physiol. 2012;303:L295-L303. https://
000337460 doi.org/10.1152/ajplung.00332.2011
111. Henderson J, Duffy L, Stratton R, Ford D, O'Reilly S. Metabolic 127. Hu B, Gharaee-Kermani M, Wu Z, Phan SH. Epigenetic regulation of
reprogramming of glycolysis and glutamine metabolism are key myofibroblast differentiation by DNA methylation. Am J Pathol.
events in myofibroblast transition in systemic sclerosis pathogene- 2010;177:21-28. https://doi.org/10.2353/ajpath.2010.090999
sis. Journal of Cellular and Molecular Medicine. 2020;24:14026- 128. Ghosh AK, Bhattacharyya S, Lafyatis R, et al. p300 is elevated in sys-
14038. https://doi.org/10.1111/jcmm.16013 temic sclerosis and its expression is positively regulated by TGF-β:
112. Bernard K, Logsdon NJ, Benavides GA, et al. Glutaminolysis is epigenetic feed-forward amplification of fibrosis. J Invest Dermatol.
required for transforming growth factor-β1-induced myofibroblast 2013;133:1302-1310. https://doi.org/10.1038/jid.2012.479
differentiation and activation. J Biol Chem. 2018;293:1218-1228. 129. Ligresti G, Caporarello N, Meridew JA, et al. CBX5/G9a/H3K9me-
https://doi.org/10.1074/jbc.RA117.000444 mediated gene repression is essential to fibroblast activation during
113. Gibb Andrew A, Lazaropoulos Michael P, Elrod John W. Myo- lung fibrosis. JCI Insight. 2019;5:e127111. https://doi.org/10.1172/
fibroblasts and fibrosis. Circ Res. 2020;127:427-447. https://doi. jci.insight.127111
org/10.1161/CIRCRESAHA.120.316958 130. Huang SK, Scruggs AM, Donaghy J, et al. Histone modifications are
114. Bai L, Bernard K, Tang X, et al. Glutaminolysis epigenetically regu- responsible for decreased Fas expression and apoptosis resistance
lates antiapoptotic gene expression in idiopathic pulmonary fibrosis in fibrotic lung fibroblasts. Cell Death Dis. 2013;4:e621. https://doi.
fibroblasts. Am J Respir Cell Mol Biol. 2019;60:49-57. https://doi. org/10.1038/cddis.2013.146
org/10.1165/rcmb.2018-0180OC 131. Sanders YY, Hagood JS, Liu H, Zhang W, Ambalavanan N,
115. Lombardi AA, Gibb AA, Arif E, et al. Mitochondrial calcium exchange Thannickal VJ. Histone deacetylase inhibition promotes fibroblast
links metabolism with the epigenome to control cellular differentia- apoptosis and ameliorates pulmonary fibrosis in mice. Eur Respir J.
tion. Nat Commun. 2019;10:4509. https://doi.org/10.1038/s41467- 2014;43:1448-1458. https://doi.org/10.1183/09031936.
019-12103-x 00095113
MERKT ET AL. 13

132. Glenisson W, Castronovo V, Waltregny D. Histone deacetylase 4 is 151. Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and
required for TGFbeta1-induced myofibroblastic differentiation. Bio- selective BCL-2 inhibitor, achieves antitumor activity while sparing
chim Biophys Acta. 2007;1773:1572-1582. https://doi.org/10. platelets. Nat Med. 2013;19:202-208. https://doi.org/10.1038/nm.
1016/j.bbamcr.2007.05.016 3048
133. Jafarinejad-Farsangi S, Farazmand A, Mahmoudi M, et al. MicroRNA- 152. Tse C, Shoemaker AR, Adickes J, et al. ABT-263: a potent and orally
29a induces apoptosis via increasing the Bax:Bcl-2 ratio in dermal bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68:3421-3428.
fibroblasts of patients with systemic sclerosis. Autoimmunity. 2015;48: https://doi.org/10.1158/0008-5472.CAN-07-5836
369-378. https://doi.org/10.3109/08916934.2015.1030616 153. Park CM, Oie T, Petros AM, et al. Design, synthesis, and computa-
134. Li CX, Talele NP, Boo S, et al. MicroRNA-21 preserves the fibrotic tional studies of inhibitors of Bcl-XL. J Am Chem Soc. 2006;128:
mechanical memory of mesenchymal stem cells. Nat Mater. 2017; 16206-16212. https://doi.org/10.1021/ja0650347
16:379-389. https://doi.org/10.1038/nmat4780 154. Petros AM, Dinges J, Augeri DJ, et al. Discovery of a potent inhibitor
135. Jazbutyte V, Fiedler J, Kneitz S, et al. MicroRNA-22 increases senes- of the antiapoptotic protein Bcl-xL from NMR and parallel synthesis.
cence and activates cardiac fibroblasts in the aging heart. Age J Med Chem. 2006;49:656-663. https://doi.org/10.1021/
(Dordr). 2013;35:747-762. https://doi.org/10.1007/s11357-012- jm0507532
9407-9 155. Pan J, Li D, Xu Y, et al. Inhibition of Bcl-2/xl with ABT-263 selec-
136. Hamsanathan S, Alder JK, Sellares J, Rojas M, Gurkar AU, Mora AL. tively kills senescent type II pneumocytes and reverses persistent
Cellular senescence: the Trojan horse in chronic lung diseases. pulmonary fibrosis induced by ionizing radiation in mice. Int J Radiat
Am J Respir Cell Mol Biol. 2019;61:21-30. https://doi.org/10.1165/ Oncol Biol Phys. 2017;99:353-361. https://doi.org/10.1016/j.ijrobp.
rcmb.2018-0410TR 2017.02.216
137. He S, Sharpless NE. Senescence in health and disease. Cell. 2017; 156. Hecker L, Logsdon NJ, Kurundkar D, et al. Reversal of persistent
169:1000-1011. https://doi.org/10.1016/j.cell.2017.05.015 fibrosis in aging by targeting Nox4-Nrf2 redox imbalance. Sci Transl
138. Meiners S, Eickelberg O, Konigshoff M. Hallmarks of the ageing Med. 2014;6(231):231ra247. https://doi.org/10.1126/scitranslmed.
lung. Eur Respir J. 2015;45:807-827. https://doi.org/10.1183/ 3008182
09031936.00186914 157. Hohmann MS, Habiel DM, Coelho AL, Verri WA Jr, Hogaboam CM.
139. Yanai H, Shteinberg A, Porat Z, et al. Cellular senescence-like fea- Quercetin enhances ligand-induced apoptosis in senescent IPF
tures of lung fibroblasts derived from idiopathic pulmonary fibrosis fibroblasts and reduces lung fibrosis in vivo. Am J Respir Cell Mol Biol.
patients. Aging (Albany NY). 2015;7:664-672. https://doi.org/10. 2019;60:28-40. https://doi.org/10.1165/rcmb.2017-0289OC
18632/aging.100807 158. Lehmann M, Korfei M, Mutze K, et al. Senolytic drugs target alveolar
140. Li Y, Liang J, Yang T, et al. Hyaluronan synthase 2 regulates fibro- epithelial cell function and attenuate experimental lung fibrosis
blast senescence in pulmonary fibrosis. Matrix Biol. 2016;55:35-48. ex vivo. Eur Respir J. 2017;50:1602367. https://doi.org/10.1183/
https://doi.org/10.1016/j.matbio.2016.03.004 13993003.02367-2016
141. Merkt W, Bueno M, Mora AL, Lagares D. Senotherapeutics: 159. Dookun E, Walaszczyk A, Redgrave R, et al. Clearance of senes-
targeting senescence in idiopathic pulmonary fibrosis. Semin Cell Dev cent cells during cardiac ischemia-reperfusion injury improves
Biol. 2020;101:104-110. https://doi.org/10.1016/j.semcdb.2019. recovery. Aging Cell. 2020;19:e13249. https://doi.org/10.1111/
12.008 acel.13249
142. Wiley CD, Velarde MC, Lecot P, et al. Mitochondrial dysfunction 160. Jia K, Dai Y, Liu A, et al. Senolytic agent Navitoclax inhibits angioten-
induces senescence with a distinct secretory phenotype. Cell Metab. sin II-induced heart failure in mice. J Cardiovasc Pharmacol. 2020;76:
2016;23:303-314. https://doi.org/10.1016/j.cmet.2015.11.011 452-460. https://doi.org/10.1097/FJC.0000000000000878
143. Müller KC, Welker L, Paasch K, et al. Lung fibroblasts from patients 161. Walaszczyk A, Dookun E, Redgrave R, et al. Pharmacological clear-
with emphysema show markers of senescence in vitro. Respir Res. ance of senescent cells improves survival and recovery in aged mice
2006;7:32-32. https://doi.org/10.1186/1465-9921-7-32 following acute myocardial infarction. Aging Cell. 2019;18:e12945.
144. Mendez MV, Stanley A, Park HY, Shon K, Phillips T, Menzoian JO. https://doi.org/10.1111/acel.12945
Fibroblasts cultured from venous ulcers display cellular characteris- 162. Moncsek A, al-Suraih MS, Trussoni CE, et al. Targeting senescent
tics of senescence. J Vasc Surg. 1998;28:876-883. https://doi.org/ cholangiocytes and activated fibroblasts with B-cell lymphoma-extra
10.1016/s0741-5214(98)70064-3 large inhibitors ameliorates fibrosis in multidrug resistance 2 gene
145. Raffetto JD, Mendez MV, Phillips TJ, Park HY, Menzoian JO. The knockout (Mdr2(/)) mice. Hepatology. 2018;67:247-259. https://
effect of passage number on fibroblast cellular senescence in doi.org/10.1002/hep.29464
patients with chronic venous insufficiency with and without ulcer. 163. Azad AI, Krishnan A, Troop L, et al. Targeted apoptosis of ductular
Am J Surg. 1999;178:107-112. https://doi.org/10.1016/s0002- reactive cells reduces hepatic fibrosis in a mouse model of cholesta-
9610(99)00134-8 sis. Hepatology. 2020;72:1013-1028. https://doi.org/10.1002/hep.
146. Harding KG, Moore K, Phillips TJ. Wound chronicity and fibroblast 31211
senescence—implications for treatment. Int Wound J. 2005;2:364- 164. Tanaka T, Yoshimi M, Maeyama T, Hagimoto N, Kuwano K, Hara N.
368. https://doi.org/10.1111/j.1742-4801.2005.00149.x Resistance to Fas-mediated apoptosis in human lung fibroblast. Eur Res-
147. King TE Jr, Bradford WZ, Castro-Bernardini S, et al. A phase 3 trial of pir J. 2002;20:359-368. https://doi.org/10.1183/09031936.02.
pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 00252602
2014;370:2083-2092. https://doi.org/10.1056/NEJMoa1402582 165. Frankel SK, Cosgrove GP, Cha SI, et al. TNF-alpha sensitizes normal
148. Distler O, Highland KB, Gahlemann M, et al. Nintedanib for systemic and fibrotic human lung fibroblasts to Fas-induced apoptosis.
sclerosis-associated interstitial lung disease. N Engl J Med. 2019; Am J Respir Cell Mol Biol. 2006;34:293-304. https://doi.org/10.
380:2518-2528. https://doi.org/10.1056/NEJMoa1903076 1165/rcmb.2005-0155OC
149. Richeldi L, du Bois RM, Raghu G, et al. Efficacy and safety of 166. Park JS, Oh Y, Park YJ, et al. Targeting of dermal myofibroblasts
nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014;370: through death receptor 5 arrests fibrosis in mouse models of sclero-
2071-2082. https://doi.org/10.1056/NEJMoa1402584 derma. Nat Commun. 2019;10:1128. https://doi.org/10.1038/
150. Maher TM, Strek ME. Antifibrotic therapy for idiopathic pulmonary s41467-019-09101-4
fibrosis: time to treat. Respir Res. 2019;20:205. https://doi.org/10. 167. Oh Y, Park O, Swierczewska M, et al. Systemic PEGylated TRAIL
1186/s12931-019-1161-4 treatment ameliorates liver cirrhosis in rats by eliminating activated
14 MERKT ET AL.

hepatic stellate cells. Hepatology. 2016;64:209-223. https://doi.org/ 179. Ugai K, Matsuda S, Mikami H, et al. Inhibition of the SET8 pathway
10.1002/hep.28432 ameliorates lung fibrosis even through fibroblast dedifferentiation.
168. Logtenberg MEW, Scheeren FA, Schumacher TN. The Front Mol Biosci. 2020;7:192. https://doi.org/10.3389/fmolb.2020.
CD47-SIRPalpha immune checkpoint. Immunity. 2020;52:742-752. 00192
https://doi.org/10.1016/j.immuni.2020.04.011 180. Suzuki K, Kim J‑D, Ugai K, et al. Transcriptomic changes involved in
169. Lerbs T, Cui L, King ME, et al. CD47 prevents the elimination of dis- the dedifferentiation of myofibroblasts derived from the lung of a
eased fibroblasts in scleroderma. JCI Insight. 2020;5:e140458. patient with idiopathic pulmonary fibrosis. Mol Med Rep. 2020;22:
https://doi.org/10.1172/jci.insight.140458 1518-1526. https://doi.org/10.3892/mmr.2020.11218
170. Mullard A. FDA approves first CAR T therapy. Nat Rev Drug Discov. 181. Rinkevich Y, Walmsley GG, Hu MS, et al. Skin fibrosis. Identification
2017;16:669. https://doi.org/10.1038/nrd.2017.196 and isolation of a dermal lineage with intrinsic fibrogenic potential.
171. Aghajanian H, Kimura T, Rurik JG, et al. Targeting cardiac fibrosis Science. 2015;348(6232):aaa2151. https://doi.org/10.1126/science.
with engineered T cells. Nature. 2019;573:430-433. https://doi.org/ aaa2151
10.1038/s41586-019-1546-z 182. Jiang D, Correa-Gallegos D, Christ S, et al. Two succeeding fibro-
172. Amor C, Feucht J, Leibold J, et al. Senolytic CAR T cells reverse blastic lineages drive dermal development and the transition from
senescence-associated pathologies. Nature. 2020;583:127-132. regeneration to scarring. Nat Cell Biol. 2018;20:422-431. https://
https://doi.org/10.1038/s41586-020-2403-9 doi.org/10.1038/s41556-018-0073-8
173. Conte E, Gili E, Fagone E, Fruciano M, Iemmolo M, Vancheri C. 183. Driskell RR, Lichtenberger BM, Hoste E, et al. Distinct fibroblast lin-
Effect of pirfenidone on proliferation, TGF-beta-induced eages determine dermal architecture in skin development and repair.
myofibroblast differentiation and fibrogenic activity of primary Nature. 2013;504:277-281. https://doi.org/10.1038/nature12783
human lung fibroblasts. Eur J Pharm Sci. 2014;58:13-19. https://doi. 184. Song G, Pacher M, Balakrishnan A, et al. Direct reprogramming of
org/10.1016/j.ejps.2014.02.014 hepatic myofibroblasts into hepatocytes in vivo attenuates liver
174. Rangarajan S, Kurundkar A, Kurundkar D, et al. Novel mechanisms fibrosis. Cell Stem Cell. 2016;18:797-808. https://doi.org/10.1016/j.
for the antifibrotic action of nintedanib. Am J Respir Cell Mol Biol. stem.2016.01.010
2016;54:51-59. https://doi.org/10.1165/rcmb.2014-0445OC 185. Rezvani M, Español-Suñer R, Malato Y, et al. In vivo hepatic repro-
175. Varga J, Pasche B. Transforming growth factor beta as a therapeutic gramming of myofibroblasts with AAV vectors as a therapeutic
target in systemic sclerosis. Nat Rev Rheumatol. 2009;5:200-206. strategy for liver fibrosis. Cell Stem Cell. 2016;18:809-816. https://
https://doi.org/10.1038/nrrheum.2009.26 doi.org/10.1016/j.stem.2016.05.005
176. Lagares D, García-Fernandez RA, Jiménez CL, et al. Endothelin
1 contributes to the effect of transforming growth factor beta1 on
wound repair and skin fibrosis. Arthritis Rheum. 2010;62:878-889.
https://doi.org/10.1002/art.27307 How to cite this article: Merkt W, Zhou Y, Han H, Lagares D.
177. Leask A, Abraham DJ. The role of connective tissue growth factor, a Myofibroblast fate plasticity in tissue repair and fibrosis:
multifunctional matricellular protein, in fibroblast biology. Biochem
Deactivation, apoptosis, senescence and reprogramming.
Cell Biol. 2003;81:355-363. https://doi.org/10.1139/o03-069
178. Tschumperlin DJ, Lagares D. Mechano-therapeutics: targeting mechan- Wound Rep Reg. 2021;1–14. https://doi.org/10.1111/wrr.
ical signaling in fibrosis and tumor stroma. Pharmacol Ther. 2020;212: 12952
107575. https://doi.org/10.1016/j.pharmthera.2020.107575

You might also like