ArticlePDF AvailableLiterature Review

Mechanotransduction of Mesenchymal Stem Cells and Hemodynamic Implications

Authors:

Abstract and Figures

Mesenchymal stem cells (MSCs) possess the capacity for self-renewal and multipotency. The traditional approach to manipulating MSC's fate choice predominantly relies on biochemical stimulation. Accumulating evidence also suggests the role of physical input in MSCs differentiation. Therefore, investigating mechanotransduction at the molecular level and related to tissue-specific cell functions sheds light on the responses secondary to mechanical forces. In this review, a new frontier aiming to optimize the cultural parameters was illustrated, i.e. spatial boundary condition, which recapitulates in vivo physiology and facilitates the investigations of cellular behavior. The concept of mechanical memory was additionally addressed to appreciate how MSCs store imprints from previous culture niches. Besides, different types of forces as physical stimuli were of interest based on the association with the respective signaling pathways and the differentiation outcome. The downstream mechanoreceptors and their corresponding effects were further pinpointed. The cardiovascular system or immune system may share similar mechanisms of mechanosensing and mechanotransduction; for example, resident stem cells in a vascular wall and recruited MSCs in the bloodstream experience mechanical forces such as stretch and fluid shear stress. In addition, baroreceptors or mechanosensors of endothelial cells detect changes in blood flow, pass over signals induced by mechanical stimuli and eventually maintain arterial pressure at the physiological level. These mechanosensitive receptors transduce pressure variation and regulate endothelial barrier functions. The exact signal transduction is considered context dependent but still elusive. In this review, we summarized the current evidence of how mechanical stimuli impact MSCs commitment and the underlying mechanisms. Future perspectives are anticipated to focus on the application of cardiovascular bioengineering and regenerative medicine.
Content may be subject to copyright.
© 2023 Chinese Journal of Physiology | Published by Wolters Kluwer - Medknow 55
Review Article
Background of Mechanotransduction
Mesenchymal stem cells (MSCs) are known as broblast-like
cells that harbor the potential of proliferation and multipotency.
Manipulating expansion and lineage specication of MSCs
has attracted extensive research attention during the past
decades. However, except for the DNA methylation prole,
MSCs share similar characteristics in morphology and
specic mesenchymal markers with broblasts.[1] Moreover,
MSCs harvested from different anatomical origins are
heterogeneous in nature and express distinct Hox codes, which
have been implicated to preserve positional imprints and
specify topographic dierentiation.[2] From the perspective of
mechanistic interrogation, a detailed appreciation of molecular
pathways could propel advancements in tissue engineering.
Cell fate choice has long been known to be dependent on
biochemical induction. Dierent regimens for corresponding
lineage specication are ascertained and has been widely
Mechanotransduction of Mesenchymal Stem Cells and
Hemodynamic Implications
Ting-Wei Kao1, Yi-Shiuan Liu2, Chih-Yu Yang3,4,5, Oscar Kuang-Sheng Lee3,6,7,8,9*
1Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, 2School of Medicine, National Tsing Hua University, Hsinchu, Taiwan,
3
Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, 4Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung
University, Taipei, Taiwan, 5Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, 6Stem Cell Research Center, National
Yang Ming Chiao Tung University, Taipei, Taiwan, 7Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, 8Department of Orthopedics,
China Medical University Hospital, Taichung, Taiwan, 9Center for Translational Genomics and Regenerative Medicine Research, China Medical University Hospital,
Taichung, Taiwan
Mesenchymal stem cells (MSCs) possess the capacity for self-renewal and multipotency. The traditional approach to manipulating MSC’s
fate choice predominantly relies on biochemical stimulation. Accumulating evidence also suggests the role of physical input in MSCs
dierentiation. Therefore, investigating mechanotransduction at the molecular level and related to tissue-specic cell functions sheds light
on the responses secondary to mechanical forces. In this review, a new frontier aiming to optimize the cultural parameters was illustrated,
i.e. spatial boundary condition, which recapitulates in vivo physiology and facilitates the investigations of cellular behavior. The concept of
mechanical memory was additionally addressed to appreciate how MSCs store imprints from previous culture niches. Besides, dierent types
of forces as physical stimuli were of interest based on the association with the respective signaling pathways and the dierentiation outcome.
The downstream mechanoreceptors and their corresponding eects were further pinpointed. The cardiovascular system or immune system
may share similar mechanisms of mechanosensing and mechanotransduction; for example, resident stem cells in a vascular wall and recruited
MSCs in the bloodstream experience mechanical forces such as stretch and uid shear stress. In addition, baroreceptors or mechanosensors of
endothelial cells detect changes in blood ow, pass over signals induced by mechanical stimuli and eventually maintain arterial pressure at the
physiological level. These mechanosensitive receptors transduce pressure variation and regulate endothelial barrier functions. The exact signal
transduction is considered context dependent but still elusive. In this review, we summarized the current evidence of how mechanical stimuli
impact MSCs commitment and the underlying mechanisms. Future perspectives are anticipated to focus on the application of cardiovascular
bioengineering and regenerative medicine.
Keywords: Hemodynamics, mechanical memory, mechanotransduction, mesenchymal stem cell, spatial boundary condition
Access this article online
Quick Response Code:
Website:
www.cjphysiology.org
DOI:
10.4103/cjop.CJOP-D-22-00144
*Address for correspondence: Dr. Oscar Kuang‑Sheng Lee,
School of Medicine, Institute of Clinical Medicine, National Yang Ming Chiao
Tung University, No. 155, Section 2, Li‑Nong Street, Beitou, Taipei 112,
Taiwan.
E‑mail: oscarlee9203@gmail.com
This is an open access journal, and arcles are distributed under the terms of the Creave
Commons Aribuon‑NonCommercial‑ShareAlike 4.0 License, which allows others to remix,
tweak, and build upon the work non‑commercially, as long as appropriate credit is given and
the new creaons are licensed under the idencal terms.
For reprints contact: WKHLRPMedknow_reprints@wolterskluwer.com
How to cite this article: Kao TW, Liu YS, Yang CY, Lee OK.
Mechanotransduction of mesenchymal stem cells and hemodynamic
implications. Chin J Physiol 2023;66:55-64.
Abstract
Received: 24-Nov-2022 Revised: 14-Mar-2023 Accepted: 16-Mar-2023
Published: 20-Apr-2023
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
56 Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
applied in cell culture.[3] Interestingly, studies observed
that the induction modalities were not only limited to the
pharmaceutical approach but also extended to mechanical
stimuli.[4] The concept of mechanotransduction emerged to
address the role of physical input in stem cell dierentiation.
At first, the literature investigated the effect exerted by
substrate rigidity. A stier scaold was demonstrated to bias
MSC toward osteoblast maturation, whereas a soft one was
toward adipocyte.[5] Follow-up research inquired about the
role of other cultural parameters, for example, porous size
and dimensionality, and the types of exerted force in the
dierentiation process.[6] Besides, whether and how cells
preserve previous mechanical imprints remained enigmatic.
The concept of “mechanical memory” emerged to depict such
phenomena.[7] These preliminary studies triggered the search
for mechanical responders and relevant signaling pathways
in MSCs.
The mechanisms regarding how MSCs perceive mechanical
stimuli remain unclear. For example, the altered behavior of
local MSCs residing within the vascular lumen exemplies
the eect of hemodynamic stimulation. Yet, how cells respond
to the mechanotransduction signaling from the perspective
of cardiac and vascular pathophysiology has not been
fully comprehended. Recently, cardiovascular applications
have emerged as a novel eld for developing translational
medicine. Resident stem cells were proposed to be pivotal
in the formation of blood vessels and the pathogenesis
of atherosclerosis.[8,9] In contrast, angiogenesis and the
orchestration of anti-inammatory properties were exhibited
to be mediated by MSCs.[10] Literature that demonstrated the
molecular mechanism of MSCs mechanotransduction, the
eects exerted by mechanical characteristics of cultural niches,
or the clinical implications, especially regarding hemodynamic
parameters, was revisited [Table 1]. These studies demonstrate
that the mechanical properties of the microenvironment
modulate the fate commitment of MSCs.
How stem cells respond to mechanical stimuli is an ongoing
endeavor. Petzold and Gentleman described the impact of
mechanotransduction on cell fate choice from the perspective
of embryogenesis instead of physiopathogenesis.[11] Raman
et al. elegantly summarized those factors that participated in
mechanotransductive pathways in a mechanistic fashion.[12]
In this article, we aimed to delineate the mechanotransductive
pathway of MSCs. Critical factors in cell culture and
application were further pragmatically recognized.
the MechanisM governing Mechanotransductive
signaling
In which manner the mechanical force is transmitted
to impact stem cell lacks full consensus [Figure 1].
The “force-from-lipid” addressed the role of lipids in
mechanotransduction. Such principle was proposed based on
the understanding of prokaryotic physiology. This framework
argues that exogenous force is directly transferred through
phospholipid bilayers, thereby orchestrating channel gating.
Escherichia coli small-conductance and large-conductance
mechanosensitive channels (MscS and MscL) were identied
as the mechanosensitive responders.[13] Nevertheless, dierent
signaling transductions were observed in eukaryotic organisms.
Another concept termed “force-from-lament” pinpointed the
alteration of intracellular architecture in mechanotransductive
pathways. This principle underscored the relevance of
the cytoskeleton and focal adhesion components in force
transduction. For example, diaphanous-related formin was
pinpointed to govern the arrangement of intranuclear lamin and
the cell fate commitment of marrow-derived MSCs.[14] These
two major principles constitute the backbone of intracellular
pathways for mechanotransduction.[15]
The mechanotransduction pathway is dissected into three
essential elements for better understanding. Firstly, the types
of exerted force, for example, uid shear stress, hydrostatic
pressure, and normal force loading determine which kind of
physical stimuli is imparted. The second element refers to
corresponding mechanosensors. Piezo-type mechanosensitive
ion channel component 1/2 (Piezo1/2) and transient
receptor potential cation channel subfamily M member
7 (TRPM7) are the examples proposed to be involved in signal
transduction.[16] Piezo family, as one type of calcium channel,
answers to dynamic uid shear force and aects cell migration
by releasing adenosine triphosphate for transducing signal to
downstream bone morphogenetic protein 2 (BMP2). A recent
Figure 1: Key signaling pathways in mechanotransduction. BMP: Bone
morphogenetic protein, cAMP: Cyclic adenosine monophosphate, GPCR:
G‑protein‑coupled receptor, mTOR: Mammalian target of rapamycin,
NICD: Notch intracellular domain, PI3K: Phosphoinositide 3‑kinases,
Piezo1/2: Piezo type mechanosensitive ion channel component, ROCK:
Rho‑associated protein kinase, YAP/TAZ: Yes‑associated protein
1/transcriptional co‑activator with PDZ‑binding motif.
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
57
Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
in vivo study based on a murine model also demonstrated that
Piezo1 acts as a mechanosensory of cardiomyocytes to result
in maladaptive hypertrophy in response to pressure overload
by inducing calcium inux to activate TRPM4.[17] These are
categorized as mechanosensitive channels and lament-linked
mechanosensors. Third, the mechanotransduction pathway
is initiated through the corresponding alterations of cellular
physiology. For instance, the compression and stretching of
the extracellular matrix were intertwined with neural stem
cell dierentiation.[18] Integrin and focal adhesion were also
recognized to transduce mechanical impact.[19] In addition, the
axis of downstream molecules in the cytoplasm and within
the nucleus, for example, BMP2, intracellular Runt-related
transcription factor 2 (Runx2), and transcription factor SP7,
were pinpointed to be impacted by mechanical forces and bring
about osteoblastogenesis.[20] The eect of mechanical stimuli
was further demonstrated at the epigenetic level. MicroRNA-9
and microRNA-10 were proposed to regulate force-manipulated
Runx2 expression during dierentiation.[21] Lee et al. also
demonstrated that microRNA-10a not only responds to
dierent hemodynamic forces but also interacts with retinoic
acid receptors and histone deacetylases, therein altering the
inammatory status of the endothelium at the vascular wall.[22]
In addition, the role of nuclear mechanosensing was addressed.
Through the linker of the nucleoskeleton and cytoskeleton,
the mechanical inputs are transduced into the nucleus and
alter chromatin structure as well as transcription factors.[23]
Elucidations toward the mechanism of mechanotransduction
facilitated the understanding of how MSCs respond to physical
stimuli.
spatial Boundary condition as a Mechanical
Modulator
The characteristics of cultural material determine the niche
for MSCs dierentiation. Exertion of the mechanical stimulus
also hinges on the manipulation of the local microenvironment.
Spatial boundary condition refers to the physical peculiarities
of the surroundings, and tensegrity describes the adaptation
of focal adhesion and cytoskeleton realignment. Early
studies exhibited the correlation between matrix rigidity and
MSC lineage specication,[24] possibly through remodeled
cell shape and tension of the cytoskeleton.[25] Recent
transcriptome analysis further identied the role of long
Table 1: Components of mechanotransduction signaling and their respective effects
Type Definition Role
Force
Shear force Force parallel to the luminal wall Shear force orients F-actin and β-catenin to polarize MSCs[2]
Hydrostatic pressure Force perpendicular to luminal
wall
Hydrostatic pressure translocates Smad2 into nucleus to promote ossication
of endochondrocytes[51]
Stretching force Force in line of circumferential
direction to luminal wall
Stretching force nurtured the mechanical resistance of local stem cells[54]
Spatial boundary condition
Rigidity The stiness of cell-residing
matrix
Rigidity of culture substrate governs viscoelasticity and subsequently the
dierentiation of MSCs[23]
Dimensionality Stereoscopic/attened surface for
cell culture
Three-dimensional microenvironment recapitulates in vivo condition[32]
Mechanical memory Imprinting of mechanical input
from the previous niche
YAP/TAZ and RUNX2 were proposed to store longitudinal memory and
impact lineage specication[41]
Conduction
Via lipid Through phospholipid bilayers MscS and MscL were recognized as the mechanoresponders in
force-from-lipid principle[13]
Via lament Through cytoskeleton and focal
adhesion
Diaphanous-related formin impacts fate choice of MSCs in mediation of
intracellular actin-lamin[14]
Antxr1 responses to focal adhesion and facilitates endochondral osteogenesis[52]
Mechanoreceptors and responders
Piezo1/Piezo2 Calcium channel/propioreceptor Piezo1 releases ATP to guide cell migration and interact with downstream
BMP2[17,54,56,57]
Piezo2 locating at aortic depressor nerve responds to uctuating blood pressure
and heart rate[64,65]
TRPM7 Calcium channel TRPM7 is essential for MSC survival and correlated with Osterix to guide
dierentiation[16,58-60]
Tentonin 3 Baroreceptor Tentonin 3 is prerequisite for the maintenance of action potential[66]
GPR68 Cation channel GPR68 modulates local vascular resistance in response to uidic shear
stress[67,68]
Antxr1: Anthrax toxin receptor 1, ATP: Adenosine triphosphate, BMP2: Bone morphogenetic protein 2, GPR68: G-protein-coupled receptor 68, MSC:
Mesenchymal stem cell, MscS/L: Mechanosensitive channels of small/large conductance, Piezo1/2: Piezo-type mechanosensitive ion channel component
1/2, RUNX2: Runt-related transcription factor 2, Smad2: Mothers against decapentaplegic homolog 2, TRPM7: Transient receptor potential cation channel
subfamily M member 7, YAP/TAZ: Yes-associated protein 1/transcriptional co-activator with PDZ-binding motif
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
58 Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
non-coding RNAs (lncRNAs) in reflecting the effect of
spatial boundary conditions. The lncRNAs possess specic
subcellular distribution and modulate genetic expression.
Epitomized by MSC osteogenesis, Zhang et al. proled six
lncRNAs with regulatory potential.[26] Specically, lnc00458
presents in only undierentiated human MSCs and serves as
the promoter for NANOG.[27] Lnc00458 responds to substrate
stiness and facilitates the maturation of multipotent stem cells
toward endoderm.[28] In addition, the cytoskeleton rearranges
concurrently with the course of lineage specication. Chen
et al. illustrated that the actin laments rearranged and became
thicker during osteoblast dierentiation. The intracellular
viscoelasticity converts from viscous-like to elastic-like during
osteogenic dierentiation but remains viscous-like during
adipogenesis.[29] The alterations of physical properties further
modulate downstream intracellular gene activities[30] and
exemplify the mechanical impact on cell fate choice.
Another aspect of mechanotransduction relates to
dimensionality. The conventional approach predominantly
utilizes Petri dishes for cell culture; yet, the topographical
dierence between such articial niches and physiological
conditions confounds the observation of stem cell
dierentiation. In our previous study, three-dimensional
polyacrylamide scaffolds were manufactured with
two distinct rigidities to recapitulate in vivo substrate
stiffness and interrogate the effects of intracellular
viscoelasticity.[31] Through passive microrheology, the sti
substrate was illustrated to escalate Young’s modulus and vice
versa. The three-dimensional scaolds were demonstrated
to better promote osteogenesis under concomitant chemical
induction compared to two-dimensional scaolds.[32] Gelatin
can be used to fabricate three-dimensional scaolds with
tunable pore sizes. The optimized spherical diameters were
100 and 150 μm for osteogenesis, by which α2 and α5
integrins were illustrated to transduce the mechanical signal
generated by the curvature of the scaolds.[33] Gelatin or
polyacrylamide are the most popular ingredients for scaold
fabrication, while culture materials, e.g., thixotropic gels,
were also feasible.[34] Besides, the expressions of cadherin-2
and cadherin-11 diered in aggregate and monolayer culture,
thereby linking dimensionality’s impact on cell fate choice.[35]
These results addressed the characteristics of the cultural
microenvironment modulates the mechanotransduction.
effect of Mechanical MeMory
The longitudinal eect is another parameter that could aect
MSC differentiation. Literature suggests that MSCs can
conserve the information regarding the previous cultural
niche, by which mechanically induced differentiation is
inuenced. To investigate this “mechanical memory,” Wu et al.
manufactured an elastomeric nanohybrid matrix with a stiness
thermo-responsive. The initial rigidity and subsequent
softening of substrate tuned by temperature were intertwined
with the in vitro chondrogenic dierentiation of MSCs.[36]
Literature also demonstrated that the mechanical memory of
initial culture expansion on a sti matrix compromised the
adipogenic potential of stem cells.[37] These ndings inspired
further studies on the interplay between mechanical switching
and lineage specication.
Although the exact mechanism responsible for mechanical
memory remains obscure, a mathematical model was
established for better elucidation.[38] In this network, the genetic
region orchestrating the physical imprint was delineated in
conjunction with the demonstration of the predictive capability
of MSC fate choice. Yang et al., on the other hand, considered
the “dosage” of such mechanical memory.[39] Upon cell
culturing on the sti scaold, either reversible or irreversible
activation of the osteogenic markers was observed with
dierent intensities of mechanical imprinting. Subsequent
literature endorsed the impact of the priming phase upon
terminal differentiation. Furthermore, three-dimensional
chromatin reorganization correlated with the transcriptome’s
dynamic landscape.[40] Specically, Yes-associated protein 1/
transcriptional co-activator with PDZ-binding motif (YAP/
TAZ) and Runx2 were reported to be responsible for storing
previous mechanical memory and thereby alternating
subsequent lineage specications.[41] The actin-actin bonding
and polarity of depolymerization under cyclic force were
also critical in transducing the temporal mechanical eect
on stem cell dierentiation.[42] Conjunctionally, substrate
stiness-dependent activation of Wnt/β-catenin signaling
participates in mechanotransduction. The expression of Wnt
and β-catenin is altered by extracellular matrix elasticity via
integrin and focal adhesion responses and stores mechanical
stimuli to further regulate cell fate.[43,44] As for epigenetic
modications, the mechanical stimuli were illustrated to
remodel and memorize chromatin’s architecture. Interestingly,
recent in vitro studies demonstrated that human MSCs
cultured on rigid substrates for 10 days exhibited irreversible
histone acetylation even after substrate softening.[45]
Megakaryoblastic Leukemia 1,2 gene and Serum Response
Factor were hypothesized to orchestrate the reprogramming
of dierentiation lineage by controlling the accessibility of
the genome.[46]
In addition, the duration of cell culture was considered to
inuence the dynamic changes of mechanically regulated
dierentiation. Specically, microRNA miR-21 was pinpointed
as a sensor with memory. Previous literature has recognized
miR-21 to correspond with mechanical input and guide stem
cells toward osteogenesis through the activated mothers
against decapentaplegic homolog 2 (Smad2)/Runx2 signaling
pathway.[47] Manipulating this microRNA erased previous
memory and, for further induction, achieved resensitization.[48]
These results together challenged the current culture protocol,
as stem cells are predominantly maintained and expanded
in a rigid microenvironment at a very early stage. Whether
the dierentiation potency would be preserved regarding
mechanical memory is amidst ongoing arguments.[49] Together,
the mechanical imprints alter subsequent lineage specications
of MSCs.
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
59
Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
type of force and Mechanoreceptors
To appreciate the mechanical eect on MSC proliferation and
cell fate choice, two aspects are considered: the type of forces
and their corresponding mechanosensors. The most widely
investigated mechanical input was the shear force, dened as
the mechanical input in a tangential direction with respect to
the lumen. Chen et al. proposed that the laminar shear force
guided F-actin orientation and MSC polarization, which
eventually led to cardiovascular maturation and angiogenesis.[2]
Laminar shear force against the inner endothelium, i.e. tunica
intima, was also demonstrated to promote local endothelial
cell dierentiation.[50] In addition, oscillatory shear stress was
exhibited to alter the organization of F-actin and orchestrate
β-catenin to determine lineage specication.[51] For example,
bronectin as the ligand of focal adhesions was proposed to
participate in ossication. Anthrax toxin receptor 1 (Antxr1)
was recently identied as mandatory in endochondral bone
formation.[52] Regulated by osteogenic marker Runx 2, Antxr1
participated in the mechanotransduction of bone marrow
stromal cells. Another type of force is the hydrostatic pressure
that acts perpendicular to the luminal wall. In conjunction
with shear force, these interactively bring about the nuclear
translocation of Smad2 to propel endochondral ossication.[53]
Other signicant classications included stretching force. In
line with the circumferential direction of the tubal surface, the
resulting strain was proposed to facilitate the dierentiation
and further enhance the mechanical resistance of residing skin
stem cells locally.[54]
As for other downstream mechano-responders, several genes were
pinpointed to be involved in regulating stem cell dierentiation.
First, Piezo1 has been extensively studied for its role in
mechanotransduction due to its kinetics of rapid inactivation.[55]
As a calcium channel, Piezo1 responds to external stimuli by
releasing adenosine triphosphate and inuencing the migration of
dental pulp-derived MSCs.[56] Moreover, Piezo1 was suggested
to inuence MSC dierentiation through BMP2. Sugimoto et al.
discovered that osteogenesis induced by hydrostatic pressure
was dependent on Piezo1-regulated BMP2.[57] Nonetheless,
although Piezo1 has been well demonstrated to be correlated
with mechanotransduction, whether this stretch-sensitive
baroreceptor participates in signaling transduction or acts as
a bystander warranted further validation. Second, TRPM7
is pivotal for MSC mechanotransduction in bone formation.
An early study suggested that TRPM7 is a prerequisite to
maintaining MSC viability.[58] It also addressed that TRPM7
might be involved in the dierentiation thereof. Intertwined with
the downstream BMP2/Smad/Runx2/Osterix axis, TRPM7 was
displayed to perceive intermittent uid shear stress and indicated
to promote endochondral/intramembranous ossification.[59]
An in vitro study further demonstrated that the knockdown of
TRPM7 prevented mechanically induced calcium inux and
osteogenesis in MSCs.[60]
Recently, various studies revealing molecular mechanisms of
pressure sensing in the cardiovascular system could facilitate a
better understanding of mechanosensing in cellular physiology.
An interesting topic reflecting mechanotransduction is
baroreex, i.e., the mechanism that maintains homeostasis
of blood pressure by sensing intraluminal pressure. Those
mechanoreceptors aggregating at the common carotid artery,
carotid sinus, and aortic arch sustain arterial pressure. The
autonomic activity and vascular tone are subsequently adjusted
accordingly to maintain hemodynamic homeostasis.[61]
Members of the Piezo family are prerequisites for this reex.[62]
Zeng et al. proposed that Piezo1 located on the sensory ganglion
was significant in blood pressure control through its
mechanosensing ability.[63] Min et al. employed a genetic
approach and pinpointed Piezo2 in the aortic depressor nerve
to participate in mechanotransduction.[64] The alteration of
blood pressure and heart rate was erased once the Piezo2
neurons were ablated. This was clinically signicant since
compromised baroreex has been related to an increased
risk of coronary artery disease and heart failure.[65] Another
mechanically evoked cation channel was Tentonin 3. Animal
studies demonstrated that tentonin 3 knockout interfered with
the stability of action potentials by pressure stimuli.[66] Other
clinical consequences included tachycardia and elevated mean
arterial blood pressure. Another pivotal player in vascular
physiology was G-protein coupled receptor 68 (GPR68).
As a mechanoreceptor in the endothelium, GPR68 reacted
to uid shear stress and altered local vascular resistance.
Specically, GPR68 responds to extracellular acidosis and
activates either the phospholipase C formation and the
calcium ux from the endoplasmic reticulum or the release
of adenylyl cyclase/cyclic adenosine 3’,5’-monophosphate
pathway into the cell for maintaining contractile phenotype.[67]
Rodent experiments further suggested a deciency in GPR68
prevented ow-induced vasodilation.[68] Together, these studies
highlighted how the mechanical eect aects uid dynamics.
Furthermore, whether these mechanoreceptors play similar
roles in stem cells or other types of cells is worth further
investigation.
Mechanotransduction and cardiovascular
iMplications
Based on a translational viewpoint, a detailed appreciation
of the mechanotransductive property is a prerequisite for
understanding the pathogenesis and pinpointing the therapeutic
target [Table 2]. A longitudinal analysis of publication count
highlighted the constant attention on the cardiovascular
translation of MSC manipulation.[69] First, heart failure
has been the major clinical burden among cardiovascular
etiologies. The hallmark of the pathogenesis was accounted as
remodeling maladaptation and ultimate decompensation. In the
presence of stress, for example, ischemia and inammation,
the quiescent resident broblasts would be induced for lineage
specication toward myobroblasts and thereby accumulate
collagen and excessively produce focal adhesion.[70] Enhanced
mechanical burden stiens the myocardium, which eventually
brings about scarring. Conversely, the rigidied substrate also
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
60 Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
promoted the migration and dierentiation of broblasts. The
Hippo pathway involving YAP/TAZ was considered relevant
to the mechanotransduction, as demonstrated by a murine
cell line.[71] Stiened extracellular matrix further deteriorated
cardiac pumping functions and eventually led to heart failure.
Another aspect regarding the mechanical eect on cardiac
function is valvular physiology. Embryological studies
suggested that the regurgitant ow promoted the recruitment
of endocardial cells, the morphogenesis of valve leaets, and
cardiac chamber formation by cytoskeleton rearrangement.
Endocardial-mesenchymal transition and shear force also
participated in this process.[72] Similar to the pathogenesis
of heart failure, dormant valve interstitial cells responding
to maximal shear stress were predisposed to aortic valve
calcication through the alteration of cytoskeleton mechanical
properties secondary to physical stimuli. Bouchareb et al.
pinpointed RhoA/rho-associated protein kinase as the key
factor in manipulating the migration of YAP into the nucleus
in response to mechanical stimuli on the cytoskeleton.[73] A
disease-in-a-dish model was established to recapitulate the
pathophysiological cardiac valvulogenesis.[74] Illuminated by
mitral valve prolapse, the platform identied specic genetic
defects that predisposed extracellular matrix derangement and
subsequently impacted the mechanotransduction of resident
stem cells.[75]
In addition, mechanical impact contributes to atherosclerosis.
Optimization of vascular elasticity is a prerequisite to
maintaining the normal physiology of blood vessels. The
governing factor was attributed to the fluid shear force.
Krüppel-like factor-2 was proposed as the key player
in transducing mechanical stimuli toward the vascular
endothelium.[76] Arterialization was proposed to reflect
shear-manipulated ephrinB2 gene expression. As for the
extracellular architecture, the rigidity of the local niche
was demonstrated to determine cell fate choice. Wong et al.
reported that the sti substrate bias the vascular progenitor cells
toward smooth muscle through the Notch signaling pathway,
while the soft matrix promotes endothelium formation.[77]
Remodeling of the vascular wall was also involved with
physical input, as the rearrangement of vimentin intermediate
laments responded to the exerted pulsatile force.[78] Moreover,
an inadequate adaptation of the local mechanical input brings
about pathological consequences. Metalloproteases secreted
from smooth muscle cells were pinpointed to be corresponding
with altered hemodynamics. Stem cells were also proposed
to participate in immunomodulatory eects regarding the
pathogenesis of atherosclerosis.[79]
Besides, MSCs were proposed to be mobilized from bone
marrow and participate in neoangiogenesis for vascular repair.
Vascular endothelial cells sense disturbed shear ow and
responded to the mechanical stress by initiating the transduction
signal of PKA-MAPK-Akt, Ras/PI3K/Akt pathways, as well
as the activation of the inux calcium channel. In conjunction,
integrins, vascular endothelial growth factor receptors, receptor
tyrosine kinases, and G protein-coupled receptors are the
pivotal mechanosensors that perceive the pathological input
of disturbed blood ow.[80] The endothelial progenitor cells
are eventually stimulated for induction toward the formation
of endothelium lining and smooth muscle cells. The small
Rho GTPases, RhoA, Cdc42, and Rac1 are pinpointed to alter
the transcription process.[81] Another mechanosensor was the
mammalian target of rapamycin couple 2, which is activated
by focal adhesion and rearranges the F-actin of MSCs.[82]
Furthermore, Chen et al. illustrated that laminar shear stress
facilitated β-catenin nuclear localization and changed the
polarity of MSCs to promote angiogenesis.[2] As exemplied
by the injury of the abdominal aorta, MSCs were demonstrated
to migrate from adjacent intact intima and subsequently
dierentiated into smooth muscle cells for vessel repair and
formation.[83] Future perspectives will focus on the clinical
application of stem cells in cardiovascular disease management.
Exemplied by recapitulating ischemic heart disease, priming
MSCs with oxidative stress was demonstrated to facilitate
engraftment and cardiogenic dierentiation. In conjunction,
stimulation with transforming growth factor-β, BMP-4, and
interleukin-6 further promoted the generation of “cardiopoietic”
stem cells for modeling ischemic cardiomyopathy.[84] Yet, the
clinical translation of manipulating MSCs still awaits further
validation.
future perspective and clinical translation
Further integrative investigations toward the factors
participating in mechanotransduction pathways were grounded
on emerging technologies. Computational mathematical
modeling serves as a state-of-the-art modality for analyzing
cellular behaviors at the molecular level. The molecular theory
Table 2: Mechanotransduction of stem cells correlates with the pathogenesis of cardiovascular diseases
Disease Pathogenesis in mechanical perspective Cellular responses and signaling transduction
Heart failure Maladaptative to excessive hemodynamic loading and
eventually aggravating to decompensatory remodeling
Local quiescent cells were induced dierentiation toward
myobroblast and resulted in scaring through YAP/TAZ
in the Hippo pathway[71]
Valvular heart
disease
Dysfunctional leaet, chordae, or papillary muscle leads
to nonphysiological blood ow within cardiac chambers
Regurgitant shear force activates indwelling dormant
cells to bring about valve calcication via Rho/ROCK[73]
Atherosclerosis Degeneration and stiening of vascular wall
characterized by intraluminal thickening and secondary
to overloaded luminal shear force
Fluid shear force stimulates the vascular endothelium
and interacts with extracellular matrix depending on the
mediation of Klf2, ephrinB2 and Notch signaling[76]
Klf2: Krüppel-like factor 2, ROCK: Rho-associated protein kinase, YAP/TAZ: Yes-associated protein 1/transcriptional co-activator with PDZ-binding motif
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
61
Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
of motor clutch demonstrates the eect exerted through matrix
rigidity upon force transmission.[85] Besides, to confer the
confounding mechanical input and for precise quantication
of the force applied, together with atomic force microscopy,[86]
video particle tracking microrheology was developed as well.
Intracellular viscoelasticity[87] and the mechanical responses to
external cytokines were depicted using such platform[88] during
cell dierentiation. Employing the tension sensor of Förster
resonance energy transfer to dene the force gradient[89] and
further to the assessment at the single-molecule level[90] are
advanced illustrations reecting the industrial evolvement of
experimental apparatus to analyze mechanotransduction. These
advancements propelled a more accurate appraisal of MSCs’
physiology and dierentiative behaviors.
On the clinical end, the frontier for MSC mechanotransduction
addressed the translation in cardiovascular applications.
Aside from previous experience in other elds, the advanced
understanding of the physical stimuli inspired interest in how
hemodynamics aects the physiology of local stem cells.
For instance, atherosclerosis and elevated blood pressure
intensify the mechanical loading on the vascular wall. How
the indwelling stem cells respond was elusive. Likewise, the
turbulent ow secondary to valvular regurgitation causes a
mechanical impact on the intra-cardiac stem cell.[91] An in vivo
study demonstrated that the steady, pulsatile shear stress or no
ow condition dierently altered the actin lament organization
of MSCs.[92] Recognition of such mechanotransduction propels
the manufacturing of articial heart valves for transplantation
purposes. In conjunction with the cellular construction of other
cardiac components, i.e. ventricle musculature for pumping,
future elucidations toward the respective mechanical signals
and MSCs adaptation will realize the cellular therapeutics.
potential therapeutics utilizing MesenchyMal
steM cells
Utilizing stem cells for therapeutic purposes in the
cardiovascular field is promising.[93] The rationale for
employing MSCs relies on self-renewing potential and
the potency to dierentiate into cardiomyocytes and other
lineages. Preclinical studies have interrogated the ecacy
of manipulating stem cells for attenuating ischemic injury
in the myocardium. In the PROMETHEUS (Prospective
Randomized Study of MSC Therapy in Patients Undergoing
Cardiac Surgery) trial, intramyocardial injection of autologous
stem cells was demonstrated to improve both regional and
global left ventricular function after cardiac operation. The
major mechanism underlying the benet was ameliorating
tissue fibrosis, promoting angiogenesis, and facilitating
myogenesis.[94] As for intracoronary injection of stem cells,
albeit theoretically feasible, to date there is no concrete
evidence to endorse such a therapeutic measure. Future
case-controlled studies are expected to validate its ecacy.
Another modality of MSC therapeutics is to manipulate the
local residing stem cells. Although with limited dierentiation
potential, those stem cells located at the vascular wall were
demonstrated to contribute to cardiomyocyte formation.
Hypothetically, the intima senses the vascular shear ow and
is mechanically impacted through vascular endothelial growth
factor receptor-2 receptor, phosphoinositide 3-kinase-Akt
pathway, and protein kinase C-mitogen-activated protein
kinase-extracellular signal-regulated kinase.[95] Interestingly,
Mekala et al. proposed these stem cells dwelling in
the outer layer of endothelium, i.e. adventitia, can also
specify to functional myocytes under appropriate culture
niches, possibly through mechanotransduction.[96] Eective
employment of stem cell therapy will be an innovative
approach to manage heart failure with reduced ejection
fraction as well as for vascular repair. Future in vivo studies
are mandatory to determine optimized parameters of the
cultural microenvironment for applying to cardiovascular
disease models as well as to demonstrate the ecacy and
safety of such treatments.
conclusions
Mechanotransduction plays an important role in MSC
phenotype and dierentiation. Recent studies propelled
the optimization of physical parameters regarding the
microenvironment for cell culture. Rigidity, spherical
size, and dimensionality of the microenvironment were all
proposed to impact the lineage specication. Furthermore,
“time” is also a crucial component as the previous cultural
condition is imprinted as mechanical memory and thereby
inuences subsequent fate commitments. Understanding
how to tune the dierentiation and stemness properties of
MSCs mechanically will help us to better dene the cellular
quality for clinical application. Dierent types of forces and
the corresponding mechanoreceptors have been pinpointed to
elucidate the underlying molecular mechanism. Additionally,
intertwined with two governing principles, force-from-lipid
as well as force-from-lament, and in conjunction with
biochemical stimuli, the mechanical stimuli alter cellular
physiology. In the cardiovascular system, the versatile
mechanosensing is reected in stretch-sensitive ion channels
Piezo1/Piezo2 and Tentonin 3 in baroreex, as well as shear
ow-sensitive GPR68 in small-diameter vessels. As for
clinical implications, the observation of mechanical eects
in the cardiovascular system will facilitate their manipulation
of local stem cells for bioengineering, cellular therapy, and
regenerative medicine.
Authors’ contributions
TWK, LYS, CYY, and OKL conceived and designed the study.
TWK and LYS conducted the literature review. TWK prepared
the manuscript. LYS, CYY, and OKL edited the article. The
study was under the supervision of CYY and OKL. All authors
have read and approved the nal manuscript.
Financial support and sponsorship
The authors received financial support for research
purposes from the National Science and Technology
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
62 Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
Council, Taiwan (MOST 109-2314-B-010-053-MY3;
MOST 110-2811-B-010-510; MOST 110-2321-B-A49-003;
NSTC 111-2923-B-007-001-MY3), Taipei Veterans
General Hospital (V111C-155; V111D63-003-MY2-1;
VGHUST111-G6-7-2), and the Center for Intelligent Drug
Systems and Smart Bio-devices (IDS2B) from The Featured
Areas Research Center Program within the framework of
the Higher Education Sprout Project by the Ministry of
Education, Taiwan. The funders have no role in study design,
data collection, analysis, interpretation, or manuscript writing.
Conflicts of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could
be construed as a potential conict of interest.
references
1. Soundararajan M, Kannan S. Fibroblasts and mesenchymal stem cells:
Two sides of the same coin? J Cell Physiol 2018;233:9099-109.
2. Chen WT, Hsu WT, Yen MH, Changou CA, Han CL, Chen YJ, et al.
Alteration of mesenchymal stem cells polarity by laminar shear
stimulation promoting β-catenin nuclear localization. Biomaterials
2019;190-191:1-10.
3. Liu K, Yu C, Xie M, Li K, Ding S. Chemical modulation of cell fate
in stem cell therapeutics and regenerative medicine. Cell Chem Biol
2016;23:893-916.
4. Hamilton DW, Maul TM, Vorp DA. Characterization of the response of
bone marrow-derived progenitor cells to cyclic strain: Implications for
vascular tissue-engineering applications. Tissue Eng 2004;10:361-9.
5. Zhang T, Lin S, Shao X, Shi S, Zhang Q, Xue C, et al. Regulating
osteogenesis and adipogenesis in adipose-derived stem cells
by controlling underlying substrate stiness. J Cell Physiol
2018;233:3418-28.
6. Brennan CM, Eichholz KF, Hoey DA. The eect of pore size within
brous scaolds fabricated using melt electrowriting on human bone
marrow stem cell osteogenesis. Biomed Mater 2019;14:065016.
7. Heo SJ, Thorpe SD, Driscoll TP, Duncan RL, Lee DA, Mauck RL.
Biophysical regulation of chromatin architecture instills a mechanical
memory in mesenchymal stem cells. Sci Rep 2015;5:16895.
8. Zhang L, Issa Bhaloo S, Chen T, Zhou B, Xu Q. Role of resident stem cells
in vessel formation and arteriosclerosis. Circ Res 2018;122:1608-24.
9. Li Y, Shi G, Han Y, Shang H, Li H, Liang W, et al. Therapeutic
potential of human umbilical cord mesenchymal stem cells on aortic
atherosclerotic plaque in a high-fat diet rabbit model. Stem Cell Res
Ther 2021;12:407.
10. Colicchia M, Jones DA, Beirne AM, Hussain M, Weeraman D,
Rathod K, et al. Umbilical cord-derived mesenchymal stromal cells
in cardiovascular disease: Review of preclinical and clinical data.
Cytotherapy 2019;21:1007-18.
11. Petzold J, Gentleman E. Intrinsic mechanical cues and their impact on
stem cells and embryogenesis. Front Cell Dev Biol 2021;9:761871.
12. Raman N, Imran SA, Ahmad Amin Noordin KB, Zaman WS,
Nordin F. Mechanotransduction in mesenchymal stem cells (MSCs)
dierentiation: A review. Int J Mol Sci 2022;23:4580.
13. Reddy B, Bavi N, Lu A, Park Y, Perozo E. Molecular basis of
force-from-lipids gating in the mechanosensitive channel MscS. Elife
2019;8:e50486.
14. Sankaran JS, Sen B, Dudakovic A, Paradise CR, Perdue T, Xie Z, et al.
Knockdown of formin mDia2 alters lamin B1 levels and increases
osteogenesis in stem cells. Stem Cells 2020;38:102-17.
15. Cox CD, Bavi N, Martinac B. Biophysical principles of
ion-channel-mediated mechanosensory transduction. Cell Rep
2019;29:1-12.
16. Xiao E, Chen C, Zhang Y. The mechanosensor of mesenchymal stem
cells: Mechanosensitive channel or cytoskeleton? Stem Cell Res Ther
2016;7:140.
17. Yu ZY, Gong H, Kesteven S, Guo Y, Wu J, Li JV, et al. Piezo1 is the
cardiac mechanosensor that initiates the cardiomyocyte hypertrophic
response to pressure overload in adult mice. Nat Cardiovasc Res
2022;1:577-91.
18. Arulmoli J, Pathak MM, McDonnell LP, Nourse JL, Tombola F,
Earthman JC, et al. Static stretch aects neural stem cell dierentiation
in an extracellular matrix-dependent manner. Sci Rep 2015;5:8499.
19. Vitillo L, Baxter M, Iskender B, Whiting P, Kimber SJ.
Integrin-associated focal adhesion kinase protects human embryonic
stem cells from apoptosis, detachment, and dierentiation. Stem Cell
Reports 2016;7:167-76.
20. Hosogane N, Huang Z, Rawlins BA, Liu X, Boachie-Adjei O,
Boskey AL, et al. Stromal derived factor-1 regulates bone morphogenetic
protein 2-induced osteogenic dierentiation of primary mesenchymal
stem cells. Int J Biochem Cell Biol 2010;42:1132-41.
21. Luo H, Gao H, Liu F, Qiu B. Regulation of Runx2 by microRNA-9 and
microRNA-10 modulates the osteogenic dierentiation of mesenchymal
stem cells. Int J Mol Med 2017;39:1046-52.
22. Lee DY, Lin TE, Lee CI, Zhou J, Huang YH, Lee PL, et al. MicroRNA-10a
is crucial for endothelial response to dierent ow patterns via
interaction of retinoid acid receptors and histone deacetylases. Proc Natl
Acad Sci 2017;114:2072-7.
23. Bouzid T, Kim E, Riehl BD, Esfahani AM, Rosenbohm J, Yang R, et al.
The LINC complex, mechanotransduction, and mesenchymal stem cell
function and fate. J Biol Eng 2019;13:68.
24. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs
stem cell lineage specication. Cell 2006;126:677-89.
25. McBeath R, Pirone DM, Nelson CM, Bhadriraju K, Chen CS. Cell shape,
cytoskeletal tension, and RhoA regulate stem cell lineage commitment.
Dev Cell 2004;6:483-95.
26. Zhang W, Dong R, Diao S, Du J, Fan Z, Wang F. Dierential long
noncoding RNA/mRNA expression proling and functional network
analysis during osteogenic dierentiation of human bone marrow
mesenchymal stem cells. Stem Cell Res Ther 2017;8:30.
27. Ng SY, Johnson R, Stanton LW. Human long non-coding RNAs promote
pluripotency and neuronal dierentiation by association with chromatin
modiers and transcription factors. EMBO J 2012;31:522-33.
28. Chen YF, Li YJ, Chou CH, Chiew MY, Huang HD, Ho JH, et al. Control of
matrix stiness promotes endodermal lineage specication by regulating
SMAD2/3 via lncRNA LINC00458. Sci Adv 2020;6:eaay0264.
29. Chen YQ, Liu YS, Liu YA, Wu YC, Del Álamo JC, Chiou A, et al.
Bio- chemical and physical characterizations of mesenchymal stromal
cells along the time course of directed dierentiation. Sci Rep
2016;6:31547.
30. Wang N, Tytell JD, Ingber DE. Mechanotransduction at a distance:
Mechanically coupling the extracellular matrix with the nucleus. Nat
Rev Mol Cell Biol 2009;10:75-82.
31. Kao TW, Chiou A, Lin KH, Liu YS, Lee OK. Alteration of 3D matrix
stiness regulates viscoelasticity of human mesenchymal stem cells. Int
J Mol Sci 2021;22:2441.
32. Hsieh WT, Liu YS, Lee YH, Rimando MG, Lin KH, Lee OK. Matrix
dimensionality and stiness cooperatively regulate osteogenesis of
mesenchymal stromal cells. Acta Biomater 2016;32:210-22.
33. Lo YP, Liu YS, Rimando MG, Ho JH, Lin KH, Lee OK.
Three-dimensional spherical spatial boundary conditions dierentially
regulate osteogenic dierentiation of mesenchymal stromal cells. Sci
Rep 2016;6:21253.
34. Pek YS, Wan AC, Ying JY. The eect of matrix stiness on mesenchymal
stem cell dierentiation in a 3D thixotropic gel. Biomaterials
2010;31:385-91.
35. Passanha FR, Geuens T, Konig S, van Blitterswijk CA, LaPointe VL.
Cell culture dimensionality inuences mesenchymal stem cell fate
through cadherin-2 and cadherin-11. Biomaterials 2020;254:120127.
36. Wu L, Magaz A, Wang T, Liu C, Darbyshire A, Loizidou M, et al.
Stiness memory of indirectly 3D-printed elastomer nanohybrid
regulates chondrogenesis and osteogenesis of human mesenchymal
stem cells. Biomaterials 2018;186:64-79.
37. Berger AJ, Anvari G, Bellas E. Mechanical memory impairs
adipose-derived stem cell (ASC) adipogenic capacity after long-term
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
63
Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
in vitro expansion. Cell Mol Bioeng 2021;14:397-408.
38. Peng T, Liu L, MacLean AL, Wong CW, Zhao W, Nie Q. A mathematical
model of mechanotransduction reveals how mechanical memory
regulates mesenchymal stem cell fate decisions. BMC Syst Biol
2017;11:55.
39. Yang C, Tibbitt MW, Basta L, Anseth KS. Mechanical memory and
dosing inuence stem cell fate. Nat Mater 2014;13:645-52.
40. Price CC, Mathur J, Boerckel JD, Pathak A, Shenoy VB. Dynamic
self-reinforcement of gene expression determines acquisition of cellular
mechanical memory. Biophys J 2021;120:5074-89.
41. Chuang LS, Ito Y. The multiple interactions of RUNX with the
Hippo-YAP pathway. Cells 2021;10:2925.
42. Lee H, Eskin SG, Ono S, Zhu C, McIntire LV. Force-history dependence
and cyclic mechanical reinforcement of actin laments at the single
molecular level. J Cell Sci 2019;132:jcs216911.
43. Astudillo P. Extracellular matrix stiness and Wnt/β-catenin signaling
in physiology and disease. Biochem Soc Trans 2020;48:1187-98.
44. Du J, Zu Y, Li J, Du S, Xu Y, Zhang L, et al. Extracellular matrix
stiness dictates Wnt expression through integrin pathway. Sci Rep
2016;6:20395.
45. Killaars AR, Grim JC, Walker CJ, Hushka EA, Brown TE, Anseth KS.
Extended exposure to sti microenvironments leads to persistent
chromatin remodeling in human mesenchymal stem cells. Adv
Sci (Weinh) 2019;6:1801483.
46. Hu X, Liu ZZ, Chen X, Schulz VP, Kumar A, Hartman AA, et al.
MKL1-actin pathway restricts chromatin accessibility and prevents
mature pluripotency activation. Nat Commun 2019;10:1695.
47. Frith JE, Kusuma GD, Carthew J, Li F, Cloonan N, Gomez GA, et al.
Mechanically-sensitive miRNAs bias human mesenchymal stem cell
fate via mTOR signalling. Nat Commun 2018;9:257.
48. Wei D, Liu A, Sun J, Chen S, Wu C, Zhu H, et al. Mechanics-controlled
dynamic cell niches guided osteogenic dierentiation of stem cells via
preserved cellular mechanical memory. ACS Appl Mater Interfaces
2020;12:260-74.
49. Kidoaki S. Frustrated dierentiation of mesenchymal stem cells.
Biophys Rev 2019;11:377-82.
50. Potter CM, Lao KH, Zeng L, Xu Q. Role of biomechanical forces in
stem cell vascular lineage dierentiation. Arterioscler Thromb Vasc Biol
2014;34:2184-90.
51. Kuo YC, Chang TH, Hsu WT, Zhou J, Lee HH, Hui-Chun Ho J, et al.
Oscillatory shear stress mediates directional reorganization of actin
cytoskeleton and alters dierentiation propensity of mesenchymal stem
cells. Stem Cells 2015;33:429-42.
52. Jiang Q, Qin X, Yoshida CA, Komori H, Yamana K, Ohba S, et al.
Antxr1, which is a target of Runx2, regulates chondrocyte proliferation
and apoptosis. Int J Mol Sci 2020;21:2425.
53. Cheng B, Liu Y, Zhao Y, Li Q, Liu Y, Wang J, et al. The role of anthrax
toxin protein receptor 1 as a new mechanosensor molecule and its
mechanotransduction in BMSCs under hydrostatic pressure. Sci Rep
2019;9:12642.
54. Aragona M, Sifrim A, Malfait M, Song Y, Van Herck J, Dekoninck S,
et al. Mechanisms of stretch-mediated skin expansion at single-cell
resolution. Nature 2020;584:268-73.
55. Del Mármol JI, Touhara KK, Croft G, MacKinnon R. Piezo1 forms a
slowly-inactivating mechanosensory channel in mouse embryonic stem
cells. Elife 2018;7:e33149.
56. Mousawi F, Peng H, Li J, Ponnambalam S, Roger S, Zhao H, et al.
Chemical activation of the Piezo1 channel drives mesenchymal stem
cell migration via inducing ATP release and activation of P2 receptor
purinergic signaling. Stem Cells 2020;38:410-21.
57. Sugimoto A, Miyazaki A, Kawarabayashi K, Shono M, Akazawa Y,
Hasegawa T, et al. Piezo type mechanosensitive ion channel component
1 functions as a regulator of the cell fate determination of mesenchymal
stem cells. Sci Rep 2017;7:17696.
58. Cheng H, Feng JM, Figueiredo ML, Zhang H, Nelson PL, Marigo V,
et al. Transient receptor potential melastatin type 7 channel is critical
for the survival of bone marrow derived mesenchymal stem cells. Stem
Cells Dev 2010;19:1393-403.
59. Liu YS, Liu YA, Huang CJ, Yen MH, Tseng CT, Chien S, et al.
Mechanosensitive TRPM7 mediates shear stress and modulates
osteogenic dierentiation of mesenchymal stromal cells through Osterix
pathway. Sci Rep 2015;5:16522.
60. Xiao E, Yang HQ, Gan YH, Duan DH, He LH, Guo Y, et al. Brief reports:
TRPM7 senses mechanical stimulation inducing osteogenesis in human
bone marrow mesenchymal stem cells. Stem Cells 2015;33:615-21.
61. Tank J, Diedrich A, Szczech E, Luft FC, Jordan J. Baroreex regulation
of heart rate and sympathetic vasomotor tone in women and men.
Hypertension 2005;45:1159-64.
62. Lai A, Chen YC, Cox CD, Jaworowski A, Peter K, Baratchi S. Analyzing
the shear-induced sensitization of mechanosensitive ion channel Piezo-1
in human aortic endothelial cells. J Cell Physiol 2021;236:2976-87.
63. Zeng WZ, Marshall KL, Min S, Daou I, Chapleau MW, Abboud FM,
et al. PIEZOs mediate neuronal sensing of blood pressure and the
baroreceptor reex. Science 2018;362:464-7.
64. Min S, Chang RB, Prescott SL, Beeler B, Joshi NR, Strochlic DE, et al.
Arterial baroreceptors sense blood pressure through decorated aortic
claws. Cell Rep 2019;29:2192-201.e3.
65. La Rovere MT, Pinna GD, Raczak G. Baroreex sensitivity:
Measurement and clinical implications. Ann Noninvasive Electrocardiol
2008;13:191-207.
66. Lu HJ, Nguyen TL, Hong GS, Pak S, Kim H, Kim H, et al. Tentonin 3/
TMEM150C senses blood pressure changes in the aortic arch. J Clin
Invest 2020;130:3671-83.
67. Ludwig MG, Vanek M, Guerini D, Gasser JA, Jones CE, Junker U, et al.
Proton-sensing G-protein-coupled receptors. Nature 2003;425:93-8.
68. Xu J, Mathur J, Vessières E, Hammack S, Nonomura K, Favre J, et al.
GPR68 senses ow and is essential for vascular physiology. Cell
2018;173:762-75.e16.
69. Chen C, Lou Y, Li XY, Lv ZT, Zhang LQ, Mao W. Mapping current
research and identifying hotspots on mesenchymal stem cells in
cardiovascular disease. Stem Cell Res Ther 2020;11:498.
70. Herum KM, Lunde IG, McCulloch AD, Christensen G. The soft- and
hard-heartedness of cardiac broblasts: Mechanotransduction signaling
pathways in brosis of the heart. J Clin Med 2017;6:53.
71. Mosqueira D, Pagliari S, Uto K, Ebara M, Romanazzo S,
Escobedo-Lucea C, et al. Hippo pathway eectors control cardiac
progenitor cell fate by acting as dynamic sensors of substrate mechanics
and nanostructure. ACS Nano 2014;8:2033-47.
72. Mahler GJ, Frendl CM, Cao Q, Butcher JT. Eects of shear stress
pattern and magnitude on mesenchymal transformation and invasion of
aortic valve endothelial cells. Biotechnol Bioeng 2014;111:2326-37.
73. Bouchareb R, Boulanger MC, Fournier D, Pibarot P, Messaddeq Y,
Mathieu P. Mechanical strain induces the production of spheroid
mineralized microparticles in the aortic valve through a RhoA/
ROCK-dependent mechanism. J Mol Cell Cardiol 2014;67:49-59.
74. Neri T, Hiriart E, van Vliet PP, Faure E, Norris RA, Farhat B, et al.
Human pre-valvular endocardial cells derived from pluripotent stem
cells recapitulate cardiac pathophysiological valvulogenesis. Nat
Commun 2019;10:1929.
75. Hinton RB, Yutzey KE. Heart valve structure and function in
development and disease. Annu Rev Physiol 2011;73:29-46.
76. Dekker RJ, van Soest S, Fontijn RD, Salamanca S, de Groot PG,
VanBavel E, et al. Prolonged uid shear stress induces a distinct
set of endothelial cell genes, most specically lung Krüppel-like
factor (KLF2). Blood 2002;100:1689-98.
77. Wong L, Kumar A, Gabela-Zuniga B, Chua J, Singh G, Happe CL,
et al. Substrate stiness directs diverging vascular fates. Acta Biomater
2019;96:321-9.
78. Helmke BP, Goldman RD, Davies PF. Rapid displacement of vimentin
intermediate laments in living endothelial cells exposed to ow. Circ
Res 2000;86:745-52.
79. Lin Y, Zhu W, Chen X. The involving progress of MSCs based therapy
in atherosclerosis. Stem Cell Res Ther 2020;11:216.
80. Masumura T, Yamamoto K, Shimizu N, Obi S, Ando J. Shear stress
increases expression of the arterial endothelial marker ephrinB2 in
murine ES cells via the VEGF-notch signaling pathways. Arterioscler
Thromb Vasc Biol 2009;29:2125-31.
81. Tzima E. Role of small GTPases in endothelial cytoskeletal dynamics
and the shear stress response. Circ Res 2006;98:176-85.
82. Brown MA, Wallace CS, Angelos M, Truskey GA. Characterization of
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
Kao, et al.: Interplay of forces and mesenchymal stem cells
64 Chinese Journal of Physiology ¦ Volume 66 ¦ Issue 2 ¦ March-April 2023
umbilical cord blood-derived late outgrowth endothelial progenitor cells
exposed to laminar shear stress. Tissue Eng Part A 2009;15:3575-87.
83. Thompson WR, Guilluy C, Xie Z, Sen B, Brobst KE, Yen SS, et al.
Mechanically activated Fyn utilizes mTORC2 to regulate RhoA and
adipogenesis in mesenchymal stem cells. Stem Cells 2013;31:2528-37.
84. Li Q, Wang Y, Deng Z. Pre-conditioned mesenchymal stem cells:
A better way for cell-based therapy. Stem Cell Res Ther 2013;4:63.
85. Elosegui-Artola A, Oria R, Chen Y, Kosmalska A, Pérez-González C,
Castro N, et al. Mechanical regulation of a molecular clutch denes
force transmission and transduction in response to matrix rigidity. Nat
Cell Biol 2016;18:540-8.
86. Yen MH, Chen YH, Liu YS, Lee, OK. Alteration of Young’s modulus
in mesenchymal stromal cells during osteogenesis measured by atomic
force microscopy. Biochem Biophys Res Commun 2020;526:827-32.
87. Chen YQ, Kuo CY, Wei MT, Wu K, Su PT, Huang CS, et al. Intracellular
viscoelasticity of HeLa cells during cell division studied by video
particle-tracking microrheology. J Biomed Opt 2014;19:011008.
88. Daviran M, McGlynn JA, Catalano JA, Knudsen HE, Druggan KJ,
Croland KJ, et al. Measuring the eects of cytokines on the modication
of pericellular rheology by human mesenchymal stem cells. ACS
Biomater Sci Eng 2021;7:5762-74.
89. Ringer P, Weißl A, Cost AL, Freikamp A, Sabass B, Mehlich A, et al.
Multiplexing molecular tension sensors reveals piconewton force
gradient across talin-1. Nat Methods 2017;14:1090-6.
90. Chang AC, Mekhdjian AH, Morimatsu M, Denisin AK, Pruitt BL,
Dunn AR. Single molecule force measurements in living cells reveal a
minimally tensioned integrin state. ACS Nano 2016;10:10745-52.
91. Majid QA, Fricker AT, Gregory DA, Davidenko N, Hernandez Cruz O,
Jabbour RJ, et al. Natural biomaterials for cardiac tissue engineering:
A highly biocompatible solution. Front Cardiovasc Med 2020;7:554597.
92. Castellanos G, Nasim S, Almora DM, Rath S, Ramaswamy S. Stem cell
cytoskeletal responses to pulsatile ow in heart valve tissue engineering
studies. Front Cardiovasc Med 2018;5:58.
93. Attar A, Bahmanzadegan Jahromi F, Kavousi S, Monabati A, Kazemi A.
Mesenchymal stem cell transplantation after acute myocardial infarction:
A meta-analysis of clinical trials. Stem Cell Res Ther 2021;12:600.
94. Karantalis V, DiFede DL, Gerstenblith G, Pham S, Symes J, Zambrano JP,
et al. Autologous mesenchymal stem cells produce concordant
improvements in regional function, tissue perfusion, and brotic burden
when administered to patients undergoing coronary artery bypass
grafting: The prospective randomized study of mesenchymal stem cell
therapy in patients undergoing cardiac surgery (PROMETHEUS) trial.
Circ Res 2014;114:1302-10.
95. Roux E, Bougaran P, Dufourcq P, Counhal T. Fluid shear stress
sensing by the endothelial layer. Front Physiol 2020;11:861.
96. Mekala SR, Wörsdörfer P, Bauer J, Stoll O, Wagner N, Reeh L, et al.
Generation of cardiomyocytes from vascular adventitia-resident stem
cells. Circ Res 2018;123:686-99.
[Downloaded free from http://www.cjphysiology.org on Friday, April 21, 2023, IP: 24.160.119.28]
... Physical factors can be categorized into two aspects: one pertains to the physical characteristics of internal microenvironment, such as substrate stiffness and topography, [26][27][28][29] while the other involves external physical stimulations, including mechanical strain, ultrasound, laser, electrical stimulation and other stimuli. [30][31][32][33][34] Although intense physical stimulation is a major factor contributing to tissue damage, 35,36 it can be widely used as a therapeutic approach in a variety of diseases if the parameters are properly adjusted. [37][38][39][40] Previous studies have demonstrated that exosomes are highly sensitive to physical factors. ...
Article
Full-text available
Mesenchymal stem cell‐derived exosomes (MSC‐Exo) offer promising therapeutic potential for various refractory diseases, presenting a novel therapeutic strategy. However, their clinical application encounters several obstacles, including low natural secretion, uncontrolled biological functions and inherent heterogeneity. On the one hand, physical stimuli can mimic the microenvironment dynamics where MSC‐Exo reside. These factors influence not only their secretion but also, significantly, their biological efficacy. Moreover, physical factors can also serve as techniques for engineering exosomes. Therefore, the realm of physical factors assumes a crucial role in modifying MSC‐Exo, ultimately facilitating their clinical translation. This review focuses on the research progress in applying physical factors to MSC‐Exo, encompassing ultrasound, electrical stimulation, light irradiation, intrinsic physical properties, ionizing radiation, magnetic field, mechanical forces and temperature. We also discuss the current status and potential of physical stimuli‐affected MSC‐Exo in clinical applications. Furthermore, we address the limitations of recent studies in this field. Based on this, this review provides novel insights to advance the refinement of MSC‐Exo as a therapeutic approach in regenerative medicine.
... Furthermore, mouse bone marrow MSC with Piezo-channel loss demonstrated the inhibition of osteoblast differentiation because of a reduction of YAP and β-catenin [138]. The effect of mechanosensitive channels on stem cell behavior is disclosed in more detail in reviews [139,140]. ...
Article
Full-text available
The extracellular matrix (ECM) is a crucial component of the stem cell microenvironment, or stem-cell niches, and contributes to the regulation of cell behavior and fate. Accumulating evidence indicates that different types of stem cells possess a large variety of molecules responsible for interactions with the ECM, mediating specific epigenetic rearrangements and corresponding changes in transcriptome profile. Signals from the ECM are crucial at all stages of ontogenesis, including embryonic and postnatal development, as well as tissue renewal and repair. The ECM could regulate stem cell transition from a quiescent state to readiness to perceive the signals of differentiation induction (competence) and the transition between different stages of differentiation (commitment). Currently, to unveil the complex networks of cellular signaling from the ECM, multiple approaches including screening methods, the analysis of the cell matrixome, and the creation of predictive networks of protein–protein interactions based on experimental data are used. In this review, we consider the existing evidence regarded the contribution of ECM-induced intracellular signaling pathways into the regulation of stem cell differentiation focusing on mesenchymal stem/stromal cells (MSCs) as well-studied type of postnatal stem cells totally depended on signals from ECM. Furthermore, we propose a system biology-based approach for the prediction of ECM-mediated signal transduction pathways in target cells.
Article
Full-text available
Customizable manufacturing of ex vivo cell engineering is driven by the need for innovations in the biomedical field and holds substantial potential for addressing current therapeutic challenges; but it is still only in its infancy. Micro‐ and nanoscale‐engineered materials are increasingly used to control core cell‐level functions in cellular engineering. By reprogramming or redirecting targeted cells for extremely precise functions, these advanced materials offer new possibilities. This influences the modularity of cell reprogramming and reengineering, making these materials part of versatile and emerging technologies. Here, the roles of micro‐ and nanoscale materials in cell engineering are highlighted, demonstrating how they can be adaptively controlled to regulate cellular reprogramming and core cell‐level functions, including differentiation, proliferation, adhesion, user‐defined gene expression, and epigenetic changes. The current reprogramming routes used to achieve pluripotency from somatic cells and the significant potential of induced pluripotent stem cell technology for translational biomedical research are covered. Recent advances in nonviral intracellular delivery modalities for cell reprogramming and their constraints are evaluated. This paper focuses on emerging physical and combinatorial approaches of intracellular delivery for cell engineering, revealing the capabilities and limitations of these routes. It is showcased how these programmable materials are continually being explored as customizable tools for inducing biophysical stimulation. Harnessing the power of micro‐ and nanoscale‐engineered materials will be a step change in the design of cell engineering, producing a suite of powerful tools for addressing potential future challenges in therapeutic cell engineering.
Article
Full-text available
Pressure overload-induced cardiac hypertrophy is a maladaptive response with poor outcomes and limited treatment options. The transient receptor potential melastatin 4 (TRPM4) ion channel is key to activation of a Ca2+/calmodulin-dependent kinase II (CaMKII)-reliant hypertrophic signaling pathway after pressure overload, but TRPM4 is neither stretch-activated nor Ca2+-permeable. Here we show that Piezo1, which is both stretch-activated and Ca2+-permeable, is the mechanosensor that transduces increased myocardial forces into the chemical signal that initiates hypertrophic signaling via a close physical interaction with TRPM4. Cardiomyocyte-specific deletion of Piezo1 in adult mice prevented activation of CaMKII and inhibited the hypertrophic response: residual hypertrophy was associated with calcineurin activation in the absence of its usual inhibition by activated CaMKII. Piezo1 deletion prevented upregulation of the sodium–calcium exchanger and changes in other Ca2+ handling proteins after pressure overload. These findings establish Piezo1 as the cardiomyocyte mechanosensor that instigates the maladaptive hypertrophic response to pressure overload, and as a potential therapeutic target. Yu et al. show that Piezo1 is the stretch-activated mechanosensor that provides the calcium source to activate TRPM4 and the downstream CaMKII-HDAC4-MEF2 pathway, a key mediator in the cardiomyocytes’ hypertrophic response in pressure overload models.
Article
Full-text available
Mechanotransduction is the process by which physical force is converted into a biochemical signal that is used in development and physiology; meanwhile, it is intended for the ability of cells to sense and respond to mechanical forces by activating intracellular signals transduction pathways and the relative phenotypic adaptation. It encompasses the role of mechanical stimuli for developmental, morphological characteristics, and biological processes in different organs; the response of cells to mechanically induced force is now also emerging as a major determinant of disease. Due to fluid shear stress caused by blood flowing tangentially across the lumen surface, cells of the cardiovascular system are typically exposed to a variety of mechanotransduction. In the body, tissues are continuously exposed to physical forces ranging from compression to strain, which is caused by fluid pressure and compressive forces. Only lately, though, has the importance of how forces shape stem cell differentiation into lineage-committed cells and how mechanical forces can cause or exacerbate disease besides organizing cells into tissues been acknowledged. Mesenchymal stem cells (MSCs) are potent mediators of cardiac repair which can secret a large array of soluble factors that have been shown to play a huge role in tissue repair. Differentiation of MSCs is required to regulate mechanical factors such as fluid shear stress, mechanical strain, and the rigidity of the extracellular matrix through various signaling pathways for their use in regenerative medicine. In the present review, we highlighted mechanical influences on the differentiation of MSCs and the general factors involved in MSCs differentiation. The purpose of this study is to demonstrate the progress that has been achieved in understanding how MSCs perceive and react to their mechanical environment, as well as to highlight areas where more research has been performed in previous studies to fill in the gaps.
Article
Full-text available
Background Trials investigating the role of mesenchymal stem cells (MSCs) in increasing ejection fraction (LVEF) after acute myocardial infarction (AMI) have raised some controversies. This study was conducted to find whether transplantation of MSCs after AMI can help improve myocardial performance indices or clinical outcomes. Methods Randomized trials which evaluated transplantation of MSCs after AMI were enrolled. The primary outcome was LVEF change. We also assessed the role of cell origin, cell number, transplantation time interval after AMI, and route of cell delivery on the primary outcome. Results Thirteen trials including 956 patients (468 and 488 in the intervention and control arms) were enrolled. After excluding the biased data, LVEF was significantly increased compared to the baseline among those who received MSC (WMD = 3.78%, 95% CI: 2.14 to 5.42, p < 0.001, I² = 90.2%) with more pronounced effect if the transplantation occurred within the first week after AMI (MD = 5.74%, 95%CI: 4.297 to 7.183; I² = 79.2% p < 0.001). The efficacy of trans-endocardial injection was similar to that of intracoronary infusion (4% [95%CI: 2.741 to 5.259, p < 0.001] vs. 3.565% [95%CI: 1.912 to 5.218, p < 0.001], respectively). MSC doses of lower and higher than 10⁷ cells did not improve LVEF differently (5.24% [95%CI: 2.06 to 8.82, p = 0.001] vs. 3.19% [95%CI: 0.17 to 6.12, p = 0.04], respectively). Conclusion Transplantation of MSCs after AMI significantly increases LVEF, showing a higher efficacy if done in the first week. Further clinical studies should be conducted to investigate long-term clinical outcomes such as heart failure and cardiovascular mortality.
Article
Full-text available
Although understanding how soluble cues direct cellular processes revolutionised the study of cell biology in the second half of the 20th century, over the last two decades, new insights into how mechanical cues similarly impact cell fate decisions has gained momentum. During development, extrinsic cues such as fluid flow, shear stress and compressive forces are essential for normal embryogenesis to proceed. Indeed, both adult and embryonic stem cells can respond to applied forces, but they can also detect intrinsic mechanical cues from their surrounding environment, such as the stiffness of the extracellular matrix, which impacts differentiation and morphogenesis. Cells can detect changes in their mechanical environment using cell surface receptors such as integrins and focal adhesions. Moreover, dynamic rearrangements of the cytoskeleton have been identified as a key means by which forces are transmitted from the extracellular matrix to the cell and vice versa. Although we have some understanding of the downstream mechanisms whereby mechanical cues are translated into changes in cell behaviour, many of the signalling pathways remain to be defined. This review discusses the importance of intrinsic mechanical cues on adult cell fate decisions, the emerging roles of cell surface mechano-sensors and the cytoskeleton in enabling cells to sense its microenvironment, and the role of intracellular signalling in translating mechanical cues into transcriptional outputs. In addition, the contribution of mechanical cues to fundamental processes during embryogenesis such as apical constriction and convergent extension is discussed. The continued development of tools to measure the biomechanical properties of soft tissues in vivo is likely to uncover currently underestimated contributions of these cues to adult stem cell fate decisions and embryogenesis, and may inform on regenerative strategies for tissue repair.
Article
Full-text available
The Hippo–YAP signaling pathway serves roles in cell proliferation, stem cell renewal/maintenance, differentiation and apoptosis. Many of its functions are central to early development, adult tissue repair/regeneration and not surprisingly, tumorigenesis and metastasis. The Hippo pathway represses the activity of YAP and paralog TAZ by modulating cell proliferation and promoting differentiation to maintain tissue homeostasis and proper organ size. Similarly, master regulators of development RUNX transcription factors have been shown to play critical roles in proliferation, differentiation, apoptosis and cell fate determination. In this review, we discuss the multiple interactions of RUNX with the Hippo–YAP pathway, their shared collaborators in Wnt, TGFβ, MYC and RB pathways, and their overlapping functions in development and tumorigenesis.
Article
Full-text available
Background Atherosclerosis (AS) is a complex disease caused in part by dyslipidemia and chronic inflammation. AS is associated with serious cardiovascular disease and remains the leading cause of mortality worldwide. Mesenchymal stem cells (MSCs) have evolved as an attractive therapeutic agent in various diseases including AS. Human umbilical cord MSCs (UCSCs) have been used in cell therapy trials due to their ability to differentiate and proliferate. The present study aimed to investigate the effect of UCSCs treatment on atherosclerotic plaque formation and the progression of lesions in a high-fat diet rabbit model. Methods Rabbits were fed a high-fat diet and then randomly divided into three groups: control, model, and treatment groups. Rabbits in the treatment group were injected with UCSCs (6 × 10 ⁶ in 500 μL phosphate buffered saline) after 1 month of high-fat diet, once every 2 weeks, for 3 months. The model group was given PBS only. We analyzed serum biomarkers, used ultrasound and histopathology to detect arterial plaques and laser Doppler imaging to measure peripheral blood vessel blood filling, and analyzed the intestinal flora and metabolism. Results Histological analysis showed that the aortic plaque area was significantly reduced in the treatment group. We also found a significant decrease in macrophage accumulation and apoptosis, an increase in expression of scavenger receptors CD36 and SRA1, a decrease in uptake of modified low–density protein (ox-LDL), and a decrease in levels of pro-inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-α following UCSCs treatment. We also found that anti-inflammatory cytokines IL-10 and transforming growth factor (TGF)-β expression increased in the aorta atherosclerotic plaque of the treatment group. UCSCs treatment improved the early peripheral blood filling, reduced the serum lipid level, and inhibited inflammation progression by regulating the intestinal flora dysbiosis caused by the high-fat diet. More specifically, levels of the microbiota-dependent metabolite trimethylamine- N -oxide (TMAO) were down-regulated in the treatment group. Conclusions UCSCs treatment alleviated atherosclerotic plaque burden by reducing inflammation, regulating the intestinal flora and TMAO levels, and repairing the damaged endothelium.
Article
Full-text available
Human mesenchymal stem cells (hMSCs) possess potential of bone formation and were proposed as ideal material against osteoporosis. Although interrogation of directing effect on lineage specification by physical cues has been proposed, how mechanical stimulation impacts intracellular viscoelasticity during osteogenesis remained enigmatic. Cyto-friendly 3D matrix was prepared with polyacrylamide and conjugated fibronectin. The hMSCs were injected with fluorescent beads and chemically-induced toward osteogenesis. The mechanical properties were assessed using video particle tracking microrheology. Inverted epifluorescence microscope was exploited to capture the Brownian trajectory of hMSCs. Mean square displacement was calculated and transformed into intracellular viscoelasticity. Two different stiffness of microspheres (12 kPa, 1 kPa) were established. A total of 45 cells were assessed. hMSCs possessed equivalent mechanical traits initially in the first week, while cells cultured in rigid matrix displayed significant elevation over elastic (G′) and viscous moduli (G″) on day 7 (p < 0.01) and 14 (p < 0.01). However, after two weeks, soft niches no longer stiffened hMSCs, whereas the effect by rigid substrates was consistently during the entire differentiation course. Stiffness of matrix impacted the viscoelasticity of hMSCs. Detailed recognition of how microenvironment impacts mechanical properties and differentiation of hMSCs will facilitate the advancement of tissue engineering and regenerative medicine.
Article
Mechanotransduction describes activation of gene expression by changes in the cell's physical microenvironment. Recent experiments show that mechanotransduction can lead to long-term "mechanical memory", where cells cultured on stiff substrates for sufficient time (priming phase) maintain altered phenotype after switching to soft substrates (dissipation phase), as compared to unprimed controls. The timescale of memory acquisition and retention is orders of magnitude larger than the timescale of mechanosensitive cellular signaling, and memory retention time changes continuously with priming time. We develop a model that captures these features by accounting for positive reinforcement in mechanical signaling. The sensitivity of reinforcement represents the dynamic transcriptional state of the cell composed of protein lifetimes and 3D chromatin organization. Our model provides a single framework connecting microenvironment mechanical history to cellular outcomes ranging from no memory to terminal differentiation. Predicting cellular memory of environmental changes can help engineer cellular dynamics through changes in culture environments.
Article
IntroductionAdipose derived stem cells (ASCs) hold great promise for clinical applications such as soft tissue regeneration and for in vitro tissue models and are notably easy to derive in large numbers. Specifically, ASCs provide an advantage for in vitro models of adipose tissue, where they can be employed as tissue specific cells and for patient specific models. However, ASC in vitro expansion may unintentionally reduce adipogenic capacity due to the stiffness of tissue culture plastic (TCPS).Methods Here, we expanded freshly isolated ASCs on soft and stiff substrates for 4 passages before adipogenic differentiation. At the last passage we swapped the substrate from stiff to soft, or soft to stiff to determine if short term exposure to a different substrate altered adipogenic capacity.ResultsExpansion on stiff substrates reduced adipogenic capacity by 50% which was not rescued by swapping to a soft substrate for the last passage. Stiff substrates had greater nuclear area and gene expression of nesprin-2, a protein that mediates the tension of the nuclear envelope by tethering it to the actin cytoskeleton. Upon swapping to a soft substrate, the nuclear area was reduced but nesprin-2 levels did not fully recover, which differentially regulated cell commitment transcriptional factors.Conclusion Therefore, in vitro expansion on stiff substrates must be carefully considered when the end-goal of the expansion is for adipose tissue or soft tissue applications.