Sie sind auf Seite 1von 11

Received: 28 October 2017 | Accepted: 30 March 2018

DOI: 10.1002/jcp.26637

MINI-REVIEW

Magnetotherapy: The quest for tendon regeneration

Tamagno Pesqueira1,2 | Raquel Costa-Almeida1,2 | Manuela E. Gomes1,2,3

1 3B's Research Group − Biomaterials,


Biodegradables and Biomimetics, University of Tendons are mechanosensitive tissues that connect and transmit the forces generated
Minho, Headquarters of the European
by muscles to bones by allowing the conversion of mechanical input into biochemical
Institute of Excellence on Tissue Engineering
and Regenerative Medicine, Zona Industrial da signals. These physical forces perform the fundamental work of preserving tendon
Gandra, Barco, Guimarães, Portugal
homeostasis assuring body movements. However, overloading causes tissue injuries,
2 ICVS/3B's − PT Government Associate
Laboratory, Guimarães, Portugal
which leads us to the field of tendon regeneration. Recently published reviews have
3 The Discoveries Centre for Regenerative and broadly shown the use of biomaterials and different strategies to attain tendon
Precision Medicine, Headquarters at regeneration. In this review, our focus is the use of magnetic fields as an alternative
University of Minho, Barco, Guimarães,
Portugal therapy, which has demonstrated clinical relevance in tendon medicine because of
their ability to modulate cell fate. Yet the underlying cellular and molecular mechanisms
Correspondence
Manuela E. Gomes, 3B's Research Group − still need to be elucidated. While providing a brief outlook about specific signalling
Biomaterials, Biodegradables and Biomimetics,
pathways and intracellular messengers as framework in play by tendon cells,
University of Minho, Headquarters of the
European Institute of Excellence on Tissue application of magnetic fields as a subcategory of physical forces is explored, opening
Engineering and Regenerative Medicine,
up a compelling avenue to enhance tendon regeneration. We outline here useful
Avepark − Parque de Ciência e Tecnologia,
Zona Industrial da Gandra, 4805–017 Barco, insights on the effects of magnetic fields both at in vitro and in vivo levels, particularly
Guimarães, Portugal.
on the expression of tendon genes and inflammatory cytokines, ultimately involved in
Email: megomes@dep.uminho.pt
tendon regeneration. Subsequently, the potential of using magnetically responsive
Funding information biomaterials in tendon tissue engineering is highlighted and future directions in
Fundação para a Ciência e a Tecnologia,
Grant numbers: IF/00593/2015, SFRH/BD/ magnetotherapy are discussed.
96593/2013
KEYWORDS
contact-free technology, electromagnetic field, magnetic biomaterials, mechano-responsive
tissue, mechanotransduction, tendon

Abbreviations: AC, alternating current; AR, adenosine receptors; BMP, bone morphoge- 1 | SNAPSHOT ON TENDON PHYSIOL OGY
netic proteins; C3H10T1/2, mouse mesenchymal stem cell line; CaM, calmodulin; CMF,
combined magnetic field; COL1A1, Type I Collagen alpha 1; COL1A2, Type I Collagen
alpha 2; COL3A1, Type III Collagen alpha 1; DC, direct current; DCN, decorin; ECM,
Tendons, as well as ligaments, together with bone, cartilage, and
extracellular matrix; EGR, early growth response; FMOD, fibromodulin; hFOB1.19, human muscle, are all components of the musculoskeletal system. Among
fetal osteoblastic cell line; IBSP, bone sialoprotein; IL, interleukins; KLF4, krueppel-like
these, tendons act as a mechanically active bridge, being responsible
factor 4; MNP, magnetic nanoparticles; MKX, mohawk; MSCs, mesenchymal stromal cells;
NO, nitric oxide; Oct4, octomer-binding transcription factor 4; PCL, Poly(ε-caprolactone); for connecting and transmitting forces between muscles or from
PEMF, pulsed electromagnetic field; PMF, pulsed magnetic field; PDGF, platelet-derived
muscles to bones, generating motion and allowing movement of body
growth factor; PGE2, prostaglandin E synthase 2; SCX, scleraxis; SPP1, osteopontin; TCPs,
tendon stem/progenitor cells; T/C-28a2, human chondrocyte cell line; TIMP, tissue parts. Morphologically, these mechanoresponsive connective tissues
inhibitor of metalloproteinases; TGF, transforming growth factor; TNF, tumor necrosis are composed of uniaxially oriented fibres following a specific
factor; TNC, tenascin-C; TNMD, tenomodulin; VEGF, vascular endothelial growth factor;
VGCC, voltage-gated calcium channel. hierarchy, from collagen fibrils (50–500 nm in diameter) to fibre

J Cell Physiol. 2018;1–11. wileyonlinelibrary.com/journal/jcp © 2018 Wiley Periodicals, Inc. | 1


2 | PESQUEIRA ET AL.

bundles (1–10 mm in diameter) wrapped by an anti-friction sheath to both in vitro and in vivo upon magnetic stimulation, subcategory of
allow tendon gliding. physical forces, are elucidated particularly as toolkit with immuno-
Tendon tissue is constituted by a mixed population of resident modulatory ability and to govern gene expression. Lastly, instructive
cells that includes mature tenocytes and tendon stem/progenitor cells magnetically responsive biomaterials as a novel strategy toward
(TSPCs) embedded in a protein-rich niche (Bi et al., 2007). In this bioengineering tendon constructs, as well as future steps into the field
tendon niche, extracellular matrix (ECM) contains primarily fibrillar of magnetotherapy and tendon tissue engineering are discussed.
collagens (e.g., collagen type I, collagen type III), as well as
proteoglycans (e.g., decorin, biglycan, fibromodulin) and glycoproteins
2 | HO W PH Y SIC AL FO RCES REGU LAT E
(e.g., elastin, tenascin C, tenomodulin, cartilage oligomeric matrix
TEN DO N H O M E O S TAS I S
protein), and cells communicate with each other and with the
surrounding microenvironment by specific cellular membrane proteins
The delicate equilibrium between mechanical forces to which tendon
responsible for regulating cell-cell (e.g., Connexin-32, Connexin-43,
cells are exposed mediates tissue homeostasis and development of
Cadherin-11) and cell-ECM (e.g., collagen-binding integrins α1β1,
pathologies. Tendon homeostasis is underpinned by physiological
α2β1, α10β1, α11β1) interactions (Costa-Almeida et al., 2015).
levels of mechanical loading (Popov et al., 2015) while overloading
Moreover, tendon exhibits a graded morphology at both ends: on
leads to tendinopathies (Spiesz et al., 2015). In a healthy state,
one side, the tendon-to-bone insertion (enthesis) and on the other
exposing tendons to stress ≤ 25 MPa and strain ≤ 2.5%, the so-called
extremity the tendon-to-muscle (myotendinous) junction. Thus, the
“physiological levels of mechanical loading,” maintains their homeo-
singular architecture of tendons together with a harmonious interplay
stasis during daily activities (Maganaris & Paul, 1999) However,
of mechanical forces are translated into an intriguing biological
repetitive movements (wear and tear), as well as acute trauma, or
complexity and a delicately powerful functional role in the musculo-
chronic diseases may culminate in tendon injury. Thus, to attain
skeletal system. Altogether, such tissues are constantly exposed to
successful tendon regeneration it is pivotal to understand the in vivo
mechanical loading, starting by posture control (gravity) and including
microenvironment housing tissue homeostasis and healing, together
forces associated with daily activities. Therefore, understanding the
with the basic cellular and molecular mechanisms occurring in such
influence of mechanical stimuli on tendon repair and regeneration is of
mechanoresponsive tissues. In this section, we briefly highlight the role
paramount importance toward developing more effective clinical
of intracellular calcium (Ca2+) in response to mechanical stimulation
strategies to improve tissue healing and functional recovery.
and transforming growth factor (TGF)-β signalling pathway along with
Tendon injuries are of utmost concern worldwide with more than
two transcriptional factors mohawk (MKX) and early growth response
102.5 million patients in US alone suffering from injuries annually
1 (EGR1) as major players of mechanotransduction in tendon cells,
(Lomas et al., 2015). These injuries frequently take place in the Achilles
particularly at dictating gene expression.
tendon, rotator cuff, and flexor tendon, with partial or total rupture
occuring within the mid-substance or enthesis (Egger & Berkowitz,
2017; Jarvinen et al., 2001; Myer & Fowler, 2016; Thorsness & Romeo,
2.1 | Insights into tendon mechanotransduction and
2016). Upon injury, tendon undergoes a process of repair implying the
molecular effectors
formation of scars, an acellular tissue which establishes adhesions with
surrouding tissues limiting tendon gliding and motion amplitude Cells sense their surroundings by a process called mechanotransduc-
(Kannus, 2000), instead of a process of regeneration, which would lead tion, a route by which they translate mechanical forces into
to full recovery of tissue functionality without scar formation. biochemical signals, generating a biological response (Jaalouk &
Nonetheless, unsatisfatory outcomes of classical treatments implying Lammerding, 2009). Tendon cells respond to physical forces via
oral administration of anti-inflammatory drugs and/or physical activation of mechanosensitive complexes involving focal adhesion
rehabilitation, and surgical interventions lay the foundation for complex, ion channels, and second messengers interacting in an
ingenious approches with ameneable translation into the clinics. autocrine and paracrine manner (Ackermann & Hart, 2016). Calcium,
This attractive approaches range from the development of natural/ an important intracellular messenger involved in numerous cellular
synthetic substitutes (Lomas et al., 2015), instructive bioactive processes (Berridge, Lipp, & Bootman, 2000) has a transient response
molecules (Docheva, Müller, Majewski, & Evans, 2015), cell- (Gaspar, when cells are mechanically stimulated (Wall & Banes, 2005). Once
Spanoudes, Holladay, Pandit, & Zeugolis, 2015) and gene- (Tang, Zhou, generated, the Ca2+ wave propagates to the neighbouring cells via gap
Wu, Liu, & Wang, 2016) based therapies to contact-free instructive junctions (Wall & Banes, 2005). As reported in an earlier study, the
signals of magnetic- or electromagnetic-based fields, herein called concentration of intracellular Ca2+ [Ca2+]ic increased from 100 nM
magnetotherapy. (basal levels) to approximately 1,500 nM when avian tendon cells were
In this review, useful insights to attain tendon regeneration are stimulated with mechanical indentation (1 Hz for 8 hr daily during 3
discussed, starting off by exploring the importance of mechanical days) (Banes et al., 1999). A similar increase is likely to occur in a
stimulation to maintain tissue homeostasis, emphasising the signalling combinatory stimulation composed of cyclic tensile strain (4%) and
pathways and intracellular messengers involved in mechanotransduc- shear stress (1,666 μm/s) (Maeda, Hagiwara, Wang, & Ohashi, 2013).
tion. Then, the underlying cellular and molecular mechanisms in play Upon mechanical stimulation, the mechanosensing L-type voltage-
PESQUEIRA ET AL.
| 3

gated calcium channels (VGCC) have been found in tenocytes and an pathways include the TGF-β signalling (Maeda et al., 2011) and
2+
increment in [Ca ]ic levels was detected upon repetitive mechanical guanosine triphosphate Rho and Rho-associated protein kinase
stretching (1.0 Hz) in vitro in a time and magnitude-dependent manner, (ROCK) (Rho/ROCK) proteins (Xu et al., 2012), ultimately involved in
pointed as a major risk factor leading to cytoskeleton damage by the commitment of stem cells toward the tenogenic lineage (Li et al.,
disrupting actin filaments (Chen, Deng, Zhang, & Tang, 2015). Despite 2015; Xu et al., 2012). When the biochemical signals reach the nucleus,
2+
a transient response in [Ca ]ic, mechanical stimulation also leads to the transcription factors MKX, activated by transcription factor II–I
alterations in Adenosine 5′-triphosphate (ATP) and changes in gene repeat domain-containing protein 1 (gtf2ird1), and zinc finger EGR1
and protein expression (Figure 1) (Lavagnino et al., 2015). Tendon cells have been identified as molecular effectors in tendon cells (Gaut et al.,
express purinoceptors P2Y in the cell membrane which makes them 2016; Kayama et al., 2016). Emerging evidence from an in vivo study
sensitive to extracellular ATP (Tsuzaki, Bynum, Almekinders, Faber, & using the mouse model C57BL6/N demonstrated that animals
Banes, 2005) and, once stimulated, such receptors are responsible for mechanically stimulated (treadmill) for 1 hr per day 5 days a week
increasing [Ca2+]ic due to enhanced influx of ATP (Yamazaki et al., during 4 weeks, increased the expression of Mkx in comparison with
2003). For example, within physiological levels of mechanical loading, unexposed controls. On top of increased Mkx expression, an
the concentration of ATP released in human tendon cells increases upregulation of tendon-associated genes Tenomodulin (Tnmd), colla-
from 0.5–1 nM (basal levels) to 14–17 nM, which returns to basal gen type I alpha 1 (Col1a1), collagen type I alpha 2 (Col1a2) and
levels within 30 min via ecto-ATPase (Tsuzaki et al., 2005); however, fibromodulin (Fmod) was also detected; however, the expression of
elevated levels of ATP temporarily block gap junctions signalling and Scleraxis (Scx) remained similar to controls (Kayama et al., 2016). On
desensitise tendon cells to mechanical stimulation (Yamazaki et al., the other side, Scx increased in Mkx−/− mouse but no changes in
2003). In this manner, depending on the intracellular machinery that is comparison with controls were seen (Kayama et al., 2016), which
activated, tendon tissue perceives distinct stimuli differently. suggests that upon mechanical stimulation, Mkx is pivotal to modulate
At the gene level, mechanical stimulation has been demonstrated the expression of vital tendon-associated genes independently of the
to influence the expression of several tendon-associated genes, highly expression of Scx. Importantly, Mkx is required for proper tendon fibres
dictating the physiology of tendon cells. When stimulated, the signal formation with a compelling role in regulating collagen type I
propagates from the cell membrane all the way to the nucleus via a production (Ito et al., 2010). From a regenerative perspective, Mkx
harmonious nexus of signalling pathways. In tendon cells, these highly accelerates tendon stem cell differentiation while preventing

FIGURE 1 Molecular interplay of mechano-magnetic stimulated tendon-derived cells on the dynamic of Ca2+ and ATP. When cells are
mechanically stimulated, due to membrane depolarization, calcium channels enhance the intracellular concentration of calcium. Part of
intracellular calcium migrates to the endoplasmic reticulum but also calcium stores within the endoplasmic reticulum release Ca2+ into the
cytoplasm. Increased cytoplasmic calcium concentration stimulates mitochondrial oxidative metabolism, which either directly via ATP synthase
activity or indirectly via dehydrogenase activity triggers the synthesis and release of ATP. The calcium/ATP dynamics is involved in
cytoskeletal organization, protein expression profiles and balance of matrix metalloproteinases
4 | PESQUEIRA ET AL.

phenotypic drift toward chondrogenic/osteogenic lineages. (Suzuki exposed to an external PEMF (75 Hz) during 5–9 hr daily over 4 weeks
et al., 2016). showed significant improvement in the range of motion of the shoulder
Early growth response 1 is likely important for the maintenance of and a decrease in inflammation (Binder, Parr, & Hazleman, 1984).
proper tendon phenotype via expression of tendon-associated genes However, a recent study suggests that PEMF was useful only as a
Scx, Col1a1, and Col1a2, and to keep suitable mechanical properties of short-term (about 5 months) modulator of inflammation including
the tissue as demonstrated in vivo (Guerquin et al., 2013). As Egr1 reduced local inflammation, postoperative joint swelling and pain in
positively affects tendon repair and commits the differentiation of patients undergoing rotator cuff repair as no differences were
mesenchymal stem cells into the tenogenic lineage in vitro (Guerquin observed after 2 years follow-up (Osti, Buono, & Maffulli, 2015).
et al., 2013), it was demonstrated that murine pluripotent stem cells Nonetheless, how magnetotherapy modalities affect tendon resident
(C3H10T1/2) transfected to overexpress Egr1 exhibited enhanced cells still remains elusive.
expression of Scx, Tnmd, Col1a1, Col1a2 when encapsulated within a Recently, magnetic stimulation of tendon cells has re-emerged as a
3D fibrin-based system under tension (Gaut et al., 2016). In the same trend in investigating novel tissue engineering and regenerative
manner as Mkx, Egr1 expression is sensitive to mechanical stimulation medicine (TERM) approaches aiming at clinical translation toward
(tension) but strikingly, its overexpression was able to rescue the tendon regeneration. Figure 2 schematically depicts main strategies in
expression of these tendon-associated genes even under reduced TERM that take advantages of magnetic field application. In this
loading conditions (Gaut et al., 2016). Importantly to note is that Scx, regard, magnetic tissue engineering approaches involve the in vitro
Mkx, and Egr1 are all three transcription factors in play during tendon generation of mature tendon tissue constructs through the use of
development and they are involved in the tenogenic commitment of magnetically responsive biomaterials. Moreover, the translation of
stem cells improving tendon repair upon injury (Milet & Duprez, 2015). magnetotherapy into the clinics may involve (i) the application of an
Importantly, upregulation of Scx, Tnmd, Col1a1, and Col1a2 upon injury external magnetic field directly at the site of injury; (ii) the implantation
improved tendon healing in vivo in the presence of Egr1 over- of a tissue-engineered construct together with magnetic actuation; or
expressing MSCs or TSPCs, demonstrating their commitment toward (iii) the implantation of cell-free magnetically responsive biomaterials
tenogenesis (Guerquin et al., 2013; Tao, Liu, Chen, Zhou, & Tang, in combination with magnetic actuation. In this section, we explain
2015). Interestingly, the healing mechanism is likely to follow particular how magnetic stimulation directly affects the physiology of cells during
signalling branches of TGF-β pathway, partially due to active SMAD2/ an inflammatory and regenerative state, and the potential of
3 signalling in a rat model of injured Achilles tendon (EGR-1 magnetically responsive biomaterials as an instructive toolbox to
overexpressing MSCs) (Guerquin et al., 2013), and BMP12/Smad1/ guide tendon regeneration.
5/8 signalling (EGR-1 overexpressing TSPCs) in a rabbit model of
injured rotator cuff tendon (Tao et al., 2015).
3.1 | Insights of magnetic actuation dictating tendon
Altogether, these findings highlight the interplay between
cell behaviour
numerous molecular effectors and mechanical forces, which may
have a role in directing tendon regeneration. Further studies at the In a regenerative medicine scenario, the basic understanding of how an
cellular and molecular level could uncover potential mechanisms external stimulus modulates cell physiology is one of the imposed
directing tenogenic differentiation of stem cells and improving cell- challenges. At the cellular level, electromagnetic field affects cell
based therapies for tendon tissue engineering and regenerative communication, governs the cytoskeletal organisation, and structural
medicine. components of the plasma membrane and changes the dynamics of
[Ca2+]ic, as reviewed elsewhere (Pesce, Patruno, Speranza, & Reale,
2013). In particular, electromagnetic stimulation has been shown to
3 | B E Y O N D R E P A I R : SH O O T I N G FO R increase the release of the anti-inflammatory cytokine interleukin (IL)-
REGENE RATION 10, simultaneously leading to a decrease of the release of pro-
inflammatory cytokines, such as IL-6 and tumor necrosis factor (TNF-
Magnetic and electromagnetic field therapy, herein called as magneto- α), both in macrophages and lymphocytes (Vergallo et al., 2013), as well
therapy, is currently used in the clinics for the treatment of several as in tendon cells (de Girolamo et al., 2013, 2014), particularly, in a time
health conditions (e.g., diabetes, multiple sclerosis) in distinct medical and field orientation (Milovanovich et al., 2016; Ross & Harrison,
fields, given its safety and non-invasiveness characteristics (Afshari 2013), as well as field intensity and exposure period dependent
et al., 2016; Sun, Kwan, Zheng, & Cheing, 2016). As a US Food and manner (de Girolamo et al., 2014). The anti-inflammatory effect of
Drug Administration (FDA)-approved therapy, pulsed electromagnetic PEMF may somewhat rely on the fact that it is an agonist for adenosine
field (PEMF) is the most commonly used magnetotherapy modality receptors (ARs), particularly increasing the density and functionality of
(Markov, 2007). In orthopaedics, for example, PEMF positively A2A and A3 ARs seen by enhanced release of IL-10 (anti-inflammatory
modulated inflammation and reduced pain in rats suffering from cytokine) and diminished release of IL-6, TNF-α (pro-inflammatory
tendinitis and in patients recovering from arthroscopic reconstruction cytokines) in musculoskeletal tissue related cell lines (human T/C-28a2
of anterior cruciate ligament (Benazzo et al., 2008; Lee, Maffulli, Li, & chondrocytes and hFOB 1.19 osteoblasts) (Vincenzi et al., 2013).
Chan, 1997). Indeed, patients with persisted rotator cuff tendinitis Additionally, PEMF (5 Hz, 4 μT) for 90 min restored the plasma
PESQUEIRA ET AL.
| 5

FIGURE 2 Schematic illustration of magnetic tissue engineering approach involving the development of magnetically responsive
bioengineered tendon constructs via generation of mature tendon tissue in vitro. Upon clinical translation, as a contact-free therapy, the
magnetic field can be applied directly at the site of injury alone, in combination with cell-free magnetic biomaterial or bioengineered magnetic
tendon construct further modulating cell response while stimulating the tissue mechanically, therefore aiding tendon regeneration

membrane Ca2+ ATPase activity and [Ca2+]ic, subsequently inhibiting two different magnetic stimuli settings were used (magnetic flow (e.g.,
the synthesis of prostaglandin E2 (PGE2), a principal mediator of 0.25 μT up to 0.4 mT) and frequency (e.g., 7.8 Hz up to 50 Hz))
inflammation (Park, Pillinger, & Abramson, 2006), in blood lympho- regardless the time of exposure (e.g., 30 min versus 7 days of
cytes in rats suffering from rheumatoid arthritis (Selvam et al., 2007). continuous stimulation) (Denaro et al., 2011; Seeliger, Falldorf,
Because the cell membrane is considered as the main target of Sachtleben, & van Griensven, 2014). In this case, the same intracellular
electromagnetic fields (Markov, 2007), stimulation strongly influences machinery activated upon stimulation could somehow explain the
the influx of Ca2+ via volatge-gated calcium channel (VGCCs), such as similarities in cell proliferation and migration in face of distinct
the L-type calcium channel (Pall, 2013). Enhanced intracellular calcium anatomic locations from where tenocytes were harvested; however
levels together with exposure to electromagnetic field modulate the further studies are needed to elucidate this correlation fully.
activation rate of calmodulin (CaM), which, in turn, by forming the Nonetheless, the release of TGF-β and VEGF together with increased
complex Ca/CaM are involved in the dynamics of nitric oxide (NO) at migration highlight the regenerative potential of electromagnetic field
the target tissue, an essential molecule involved in the progression of stimulation of tendon tissue. Cell cycle and total collagen accumula-
tendon healing cascade (Bokhari & Murrell, 2012). For example, tion, on the other side, seem not to be influenced upon stimulation in
electromagnetic stimulation directly modulating CaM signals increased vitro neither culturing human tenocytes harvested from supraspinatus
the production of NO (2-fold in comparison with unexposed cells) in and quadriceps tendons (Denaro et al., 2011; Liu et al., 2016).
2+
human fibroblasts when high levels of [Ca ]ic were observed (Pilla, Surprisingly, TSPCs electromagnetically stimulated (30 Hz,
2012). However, these mechanisms of action are not completely 1.5 mT) for 60 min presented a similar migratory profile compared to
understood in tendon cells. Taking a step back on the anti- unexposed cells (Randelli et al., 2016). In this study, authors also
inflammatory effect of electromagnetic field by intensifying the reported that electromagnetic stimulation is likely to maintain TSPCs in
release of IL-10 in tendon cells, they also release growth (e.g., TGF- an undifferentiated state by regulating stem cell reprogramming
β) and angiogenic factors (e.g., vascular endothelial growth factor factors octamer-binding transcription factor 4 (Oct4), Kruppel-like
(VEGF)) (de Girolamo et al., 2013, 2014). Moreover, stimulated cells factor 4 (KLF4) and Nanog, up to 48 hr following stimulation (Randelli
proliferated and migrated faster than unexposed controls, even when et al., 2016). In a different study, tendon-derived cells stimulated with
6
|

TABLE 1 The influence of electromagnetic field stimulation on human tendon-derived cells in vitro
Stimulation Treatment Cell type Outcome Ref.
In vitro
PEMF (frequencies between 10 and 30 Hz Single treatment for 1 hr up to 48 hr Tendon stem/progenitor cells Significant upregulation of Oct4, KLF4, and Nanog. Randelli
and magnetic flow between 0.5–1.5 mT) cell culture from supraspinatus tendon No effects on the expression of tendon markers et al.
COL1A1 and TNC. (2016)
PEMF (frequency 75–Hz and magnetic Single treatment for 4–hr, 8 hr, and Tendon-derived cells from 8 hr treatment significantly enhances the de Girolamo
flow of 1.5 mT) 12 hr up to 48 hr cell culture semitendinous and gracilis expression of SCX (58%) and COL1A1 (49%). Release of et al.
tendons IL-6 (360%) and IL-10 (133%), (2013)
TGF-β (11-fold) and VEGF-A (41%)
PEMF (frequency 75 Hz and magnetic Single and repeated treatments for Tendon-derived cells from Repeated treatment enhances the de Girolamo
flow of 1.5 mT or 3.0 mT) 8 hr and 12 hr up to 48 hr cell semitendinous and gracilis release of IL-10 (70%) as compared with unexposed et al.
culture tendons cells (2014)
PEMF (frequency 7.8 Hz or 33 Hz, and Combined treatment for 30 min Tendon-derived cells from Enhances cell migration and proliferation Seeliger
magnetic flow of 0.25 µT or 3.16 µT) (10 min of 33 Hz plus 20 min of patellar tendon et al.
7.8 Hz) (2014)
PEMF (frequency 50 Hz and magnetic Continuous exposure for 7 days Tendon-derived cells from Enhances cell migration Denaro et al.
flow of 0.4 mT) supraspinatus and quadriceps (2011)
tendon
PEMF (Physio-Stim® PEMF system, Single treatment for 3 hr daily up to Tendon-derived cells from Increases the expression of COL1A1 (2.9-fold), Liu et al.
Orthofix Inc.) 2 weeks supraspinatus tendon TGF-β1 (3.5-fold), PDGF (6.3-fold), BMP-12 (3.0-fold), (2016)
TIMP-4 (5.1-fold) at 2 weeks under inflammatory
conditions (10 ng/ml of IL-1α)

BMP, Bone Morphogenetic Protein; COL1A1, Collagen type I; PDGF, Platelet Derived Growth Factor; SCX, Scleraxis; TIMP, Tissue Inhibitor of Metalloproteinase; TNC, Tenascin C.
PESQUEIRA
ET AL.
PESQUEIRA ET AL.
| 7

low-frequency static magnetic field (2 Hz, 350 mT) had enhanced

Hu, & Lu, 2014


Robotti, Zimbler,

Lee et al. (1997)

Xu, Zhang, Qu,


expression of COL1A1, TNC, DCN, and SCX after a single exposure for

Strauch et al.

Grossman,
Greenough

Kenna, &
8 hr (Pesqueira, Costa-Almeida, & Gomes, 2017). Subsequently, the

(1996)

(2006)

1999
expression of COL1A1, COL3A1, and TNC increased in culture upon
Ref.

magnetic stimulation for 8 hr every 24 hr up to 14 days (Pesqueira


et al., 2017). Despite the emerging evidence, it is challenging to claim
an optimal window of magnetotherapy for tendon regeneration, given
diverse cell responses observed and magnetic settings used. Major

± 1.41 MPa), energy to failure (0.87 ± 0.17 J), and a number of


Higher load to failure (311.0 ± 59.4 N), ultimate strength (8.46
outcomes of in vitro and in vivo studies with magnetic stimulation are

PMF 17 Hz showed reduced inflammation, better collagen


summarised in Tables 1 and 2, respectively. The distinct behavior may
No differences were observed compared with control

No differences were observed compared with control


Increased in tensile strength (69%) of treated tendon

be exploited by the impact that intrinsic magnetic settings such as the


magnetic field and magnetic field gradient have on intracellular forces

alignment, and fibroblasts were more mature

proteoglycans (36%) than the control group.


(Zabloskii et al., 2016) Overall, this raises an increasing attention
toward more detailed and concise studies both at the cellular and
molecular levels. As a contact-free technology and simultaneously
acting both as a mechanical stimulation and modulation of inflamma-
tory response, of particular importance for tendon regeneration,
modulate cell fate, and ultimately, direct tenogenic differentiation,
magnetic field can represent an interesting candidate to remotely
guide tissue regeneration.
Outcome

3.2 | Magnetically responsive biomaterials in


musculoskeletal tissues: The first steps in tendon
the forepaw of New Zealand

tissue engineering
Flexor digitorum profundus of

hind leg of Sprague-Dawley

Achilles tendon of Sprague-


Flexor profundus tendon of
Achilles tendon of the right

the central toe in White

Zealand White rabbits

Three-dimensional (3D) biomaterials are an essential toolbox to


Patellar tendon of New

provide physical cues, therefore orchestrating vital cell responses


Leghorn chickens

AC, alternating current; DC, direct current; CMF, combined magnetic field; PMF, pulsed magnetic field.
White Rabbits

which culminate in tissue development (Furth, Atala, & Van Dyke,


Dawley rats

2007). Among the classes of biomaterials, magnetically responsive


Cell type
The influence of electromagnetic field stimulation on tendon tissue in vivo

ones combined with on-demand magnetic stimulation are becoming an


rats

attractive strategy to regulate cell fate (Santos, Reis, & Gomes, 2015).
Such biomaterials can be fabricated through the incorporation of
Single treatment for 15 min daily for
Single treatment for 8 hr daily for 3

magnetic nanoparticles (e.g., ferromagnetic, superparamagnetic) into a


Double treatment for 30 min daily

daily up to 16 weeks following


Combined treatment for 30 min

polymeric network by simply mixing both solutions (blending method),


between Day 6 and Day 21
Single treatment for 6 hr daily

promoting in situ precipitation of magnetic precursors (in situ


third postoperative day

precipitation method) or chemically modifying the surface of nano-


particles enabling them to act as cross-linking agents (grafting-onto
for 3 weeks

method) (Li et al., 2013). Upon fabrication, the bulk properties are
Treatment

4 weeks

hooked on the production method adopted and type of nanoparticles


weeks

used (Dai & Nelson, 2010). In particular, superparamagnetic nano-


particles are promising candidates within tissue engineering strategies
given that (i) they do not retain magnetic fluctuations after removal of
± 200 mG; amphimagnetic field = ± 600 mG)
PEMF 15 Hz with 19 quasi-rectangular pulses

PMF with quasi-sinusoidal rectified waveform

the applied external magnetic field; (ii) there is reduced interparticle


magnetic interaction; and (iii) they are stable under physiological
frequency = 76.6 Hz; DC: strength =
Radio frequency PEMF of 27.12 MHz

conditions (Dai & Nelson, 2010). Bringing this knowledge together,


(17/50 Hz) PEMF (15/46 Hz)

magnetically responsive biomaterials have been reported to exert a


CMF (AC: strength = 400 mG,

positive role in bone regeneration (Singh et al., 2014). For instance,


PEMF (frequency 15 Hz)

culturing rat bone marrow MSCs on a magnetic nanofibrous scaffold


sinusoidal waves

(15% (w/v) MNPs within a polycaprolactone (PCL) mesh) resulted in an


upregulation of osteogenic genes COL1A1, osteopontin (SPP1) and
Stimulation
TABLE 2

bone sialoprotein (IBSP) (Singh et al., 2014). Additionally, in vivo


In vivo

implantation of a magnetic nanocomposite scaffold (5–10% w/w


MNPs in PCL) combined with exposure to a static magnetic field
8 | PESQUEIRA ET AL.

enhanced bone formation 6 weeks after surgery in a critical-sized frequency, magnetic flow, and magnetic gradient) and poor knowledge
defect created in mice calvaria (Yun et al., 2016). This class of of their effects on intracellular forces and machinery involved. By
biomaterials has also been reported for cartilage (Zhang, Lock, Sallee, & knowing the underlying molecular mechanisms in command upon
Liu, 2015) and muscle regeneration (Cezar et al., 2016). For example, stimulation, magnetotherapy could be an important adjuvant in tendon
the implantation of biphasic hydrogels (7% iron oxide in alginate regeneration, potentially enabling to substitute nonsteroidal anti-
covalently modified with RGD peptide) magnetically stimulated (1 Hz inflammatory drugs, for example, highly used by patients with
for 5 min every 12 hr) resulted in muscle regeneration with reduced inflammation. In this sense, future research is needed to explore (i)
fibrous capsule formation, fibrosis, and inflammation following 2 weeks how undifferentiated and differentiated cells recognize different
post-implantation in mouse defect model of tibialis anterior muscle magnetotherapy-based settings; (ii) which signalling pathways are
(Cezar et al., 2016). Also, magnetically responsive biomaterials have responsible for transducing magnetic stimuli; (iii) to what extent
been proposed for tendon tissue engineering (Gonçalves et al., 2016; magnetic stimulation can modulate tendon cell behavior in inflamma-
Santos et al., 2016; Silva et al., 2017). Indeed, protein expression of tory and non-inflammatory environment; (iv) and whether the
tendon-related ECM proteins (collagen type I and tenascin C) was expression of tendon-associated genes and proteins is altered. To
found to be increased under magnetically stimulated (2 Hz, 350 mT) conclude, we expect that contact-free magnetically stimulated 3D
adipose-derived stem cells cultured on aligned fibrous magnetic environment based approaches will advance the treatment of tendon
scaffold (0.018:1 w/w MNPs in PCL-based blend), therefore suggest- disorders and establish a novel paradigm for regenerative medicine
ing the tenogenic commitment of this cell population (Gonçalves et al., broadly.
2016). Furthermore, subcutaneous implantation of the same bioma-
terial formulation of PCL-based blend magnetically responsive
membrane (0.018:1 w/w MNPs) in a rat model has shown its ability ACKNOWLEDGMENTS
to moderate inflammation (Santos et al., 2016). The presence of M2
The authors acknowledge the financial support from FCT (Fundação
macrophages (anti-inflammatory phenotype) in the fibrous tissue was
para a Ciência e Tecnologia) in the framework of FCT-POPH-FSE for
reduced and the magnetically responsive membrane also prevented
R.C.-A. PhD grant SFRH/BD/96593/2013, and M.E.G. FCT grant IF/
mast cell infiltration and fibrous tissue thickening over time (9 weeks)
00593/2015. The authors related works were financed by the project
(Santos et al., 2016). This study is an important step to understand the
“Accelerating tissue engineering and personalized medicine discover-
immunomodulatory ability of magnetically responsive biomaterials to
ies by the integration of key enabling nanotechnologies, marine-
attain tissue regeneration, particularly because healing generally
derived biomaterials and stem cells,” supported by Norte Portugal
involves numerous cell types including inflammatory cells. The
Regional Operational Programme (NORTE 2020), under the
modulation of inflammation in early stages of tissue healing may
PORTUGAL 2020 Partnership Agreement, through the European
lead to improved healing as a result of the presence of M2
Regional Development Fund (ERDF).
macrophages in acute inflammation frequently being favorable for
immunosuppression, scar resolution, and remodeling (Thomopoulos,
Parks, Rifkin, & Derwin, 2015). AUTHOR S’ C ONT R I B UT I ON
Overall, besides acting as a physical support for cells, such
biomaterials will provide the necessary mechanical stimuli through TP and RC-A have contributed equally to literature review and
structural deformations by the application of an external magnetic manuscript preparation. All authors have read and approved the final
field, which enables the creation of a dynamic microenvironment version of the manuscript.
triggering distinct cell responses, as discussed above (Silva et al., 2017).

CONFLICTS OF INTEREST
4 | C ONC LU SI ON S
The authors have no competing interests.

Tendon physiology is intrinsically dependent on mechanical stimuli


from the moment of development onwards, highly dictating the ORCID
adaptation and survival of tendon cells. Increasing body of research has
Raquel Costa-Almeida http://orcid.org/0000-0001-7188-645X
uncovered how mechanical actuation modulates cell behavior by
Manuela E. Gomes http://orcid.org/0000-0002-2036-6291
pointing out mechanosensing molecules (Egr1, Mkx), intracellular
messengers (Ca2+, ATP) together with signaling pathways (TGF-β,
MAPK) responsible for keeping tendon cells either in the right way or
to steer the commitment of stem cells toward differentiated cells. REFERENCES
Magnetotherapy is becoming an attractive strategy in modulating cell Ackermann, P. W., & Hart, D. Editors (2016). Cell signaling in tenocytes:
physiology with promising, although junior outcomes in tendon Response to load and ligands in health and disease. Metabolic influences
healing; partially due to lack of standardized magnetic settings (e.g.,
PESQUEIRA ET AL.
| 9

on risk for tendon disorders (pp. 229–238). https://doi.org/10.1007/ Docheva, D., Müller, S. A., Majewski, M., & Evans, C. H. (2015). Biologics for
978-3-319-33943-6 tendon repair. Advanced Drug Delivery Reviews, Elsevier B.V. 84,
Afshari, D., Moradian, N., khalili, M., Razazian, N., Bostani, A., Hoseini, J., . . . 222–239. https://doi.org/10.1016/j.addr.2014.11.015
Ghiasian, M. (2016). Evaluation of pulsing magnetic field effects on Egger, A. C., & Berkowitz, M. J. (2017). Achilles tendon injuries. Current
paresthesia in multiple sclerosis patients, a randomized, double-blind, Review Musculoskeletal Medicine, 10(1), 72–80. https://doi.org/
parallel-group clinical trial. Clinical Neurology and Neurosurgery, Elsevier 10.1007/s12178-017-9386-7
B.V. 149, 171–174. https://doi.org/10.1016/j.clineuro.2016.08.015 Furth, M. E., Atala, A., & Van Dyke, M. E. (2007). Smart biomaterials design
Banes, A. J., Weinhold, P., Yang, X., Tsuzaki, M., Bynum, D., Bottlang, M., & for tissue engineering and regenerative medicine. Biomaterials, 28(34),
Brown, T. (1999). Gap junctions regulate responses of tendon cells ex 5068–5073. https://doi.org/10.1016/j.biomaterials.2007.07.042
vivo to mechanical loading. Clinical Orthopaedics and Related Research, Gaspar, D., Spanoudes, K., Holladay, C., Pandit, A., & Zeugolis, D. (2015).
367, 356–370. Progress in cell-based therapies for tendon repair. Advanced Drug
Benazzo, F., Zanon, G., Pederzini, L., Modonesi, F., Cardile, C., Falez, F., . . . Delivery Reviews, Elsevier B.V. 84, 240–256. https://doi.org/10.1016/j.
Massari, L. (2008). Effects of biophysical stimulation in patients addr.2014.11.023
undergoing arthroscopic reconstruction of anterior cruciate ligament: Gaut, L., Robert, N., Delalande, A., Bonnin, M. A., Pichon, C., & Duprez, D.
Prospective, randomized and double blind study. Knee Surgery, Sports (2016). EGR1 regulates transcription downstream of mechanical signals
Traumatology, Arthroscopy, 16(6), 595–601. https://doi.org/10.1007/ during tendon formation and healing. PLoS ONE, 11(11), 1–16. https://
s00167-008-0519-9 doi.org/10.1371/journal.pone.0166237
Berridge, M. J., Lipp, P., & Bootman, M. D. (2000). The versatility and Gonçalves, A. I., Rodrigues, M. T., Carvalho, P. P., Bañobre-López, M., Paz,
universality of calcium signalling. Nature Reviews. Molecular Cell Biology, E., Freitas, P., & Gomes, M. E. (2016). Exploring the potential of starch/
1(1), 11–21. https://doi.org/10.1038/35036035 polycaprolactone aligned magnetic responsive scaffolds for tendon
Bi, Y., Ehirchiou, D., Kilts, T. M., Inkson, C. A., Embree, M. C., Sonoyama, W., regeneration. Advanced Healthcare Materials, 5(2), 213–222. https://
. . . Young, M. F. (2007). Identification of tendon stem/progenitor cells doi.org/10.1002/adhm.201500623
and the role of the extracellular matrix in their niche. Nature Medicine, Greenough, C. (1996). The effect of pulsed electromagnetic fields on flexor
13(10), 1219–1227. https://doi.org/10.1038/nm1630 tendon healing in the rabbit. The Journal of Hand Surgery: Journal of the
Binder, A., Parr, G., & Hazleman, B. (1984). Pulsed electromagnetic field British Society for Surgery of the Hand, 21(6), 808–812. https://doi.org/
therapy of persisted rotator cuff tendinitis. A double-blind controlled 10.1016/S0266-7681(96) 80198-7
assessment. The Lancet, 1(8379), 695–698. Guerquin, M. J., Charvet, B., Nourissat, G., Havis, E., Ronsin, O., Bonnin,
Bokhari, A. R., & Murrell, G. A. C. (2012). The role of nitric oxide in tendon M. A., . . . Duprez, D. (2013). Transcription factor EGR1 directs tendon
healing. Journal of Shoulder and Elbow Surgery, Elsevier Ltd, 21(2), differentiation and promotes tendon repair. Journal of Clinical
238–244. https://doi.org/10.1016/j.jse.2011.11.001 Investigation, 123(8), 3564–3576.
Cezar, C. A., Roche, E. T., Vandenburgh, H. H., Duda, G. N., Walsh, C. J., & Ito, Y., Toriuchi, N., Yoshitaka, T., Ueno-Kudoh, H., Sato, T., Yokoyama, S.,
Mooney, D. J. (2016). Biologic-free mechanically induced muscle . . . Asahara, H. (2010). The Mohawk homeobox gene is a critical
regeneration. Proceedings of the National Academy of Sciences of the regulator of tendon differentiation. Proceedings of the National Academy
United States of America, 113(6), 1534–1539. https://doi.org/10.1073/ of Sciences of the United States of America, 107(23), 10538–10542.
pnas.1517517113 https://doi.org/10.1073/pnas.1000525107
Chen, W., Deng, Y., Zhang, J., & Tang, K. (2015). Uniaxial repetitive Jaalouk, D. E., & Lammerding, J. (2009). Mechanotransduction gone awry.
mechanical overloading induces influx of extracellular calcium and Nature Reviews. Molecular Cell Biology, 10(1), 63–73. https://doi.org/
cytoskeleton disruption in human tenocytes. Cell and Tissue Research, 10.1038/nrm2597
359(2), 577–587. https://doi.org/10.1007/s00441-014- 2018-2 Jarvinen, T. A., Kannus, P., Paavola, M., Jarvinen, T. L., Jozsa, L., & Jarvinen, M.
Costa-Almeida, R., Gonçalves, A. I., Gershovich, P., Rodrigues, M. T., Reis, (2001). Achilles tendon injuries. Current Opinion in Rheumatology, 13(2),
R. L., & Gomes, M. E. (2015). Tendon stem cell niche. In K. Turksen, (Ed.), 150–155. https://doi.org/10.1097/00002281-200103000-00009
Tissue-specific stem cell niche (pp. 221–244). First. Humana Press Kannus, P. (2000). Structure of the tendon connective tissue. Scandinavian
Springer, Cham, Switzerland. https://doi.org/10.1007/978-3-319- Journal of Medicine & Science in Sports, 10(3), 312–320. https://doi.org/
21705-5 10.1034/j.1600-0838.2000.010006312.x
Dai, Q., & Nelson, A. (2010). Magnetically-responsive self assembled Kayama, T., Mori, M., Ito, Y., Matsushima, T., Nakamichi, R., Suzuki, H., &
composites. Chemical Society Reviews, 39(11), 4057–4066. https://doi. Ichinose, S. (2016). Gtf2ird1-dependent mohawk expression regulates
org/10.1039/b812669k mechanosensing. Molecular and Cellular Biology, 36(8), 1297–1309.
de Girolamo, L., Stanco, D., Galliera, E., Viganò, M., Colombini, A., Setti, S., https://doi.org/10.1128/MCB 00950-15.Address
. . . Sansone, V. (2013). Low frequency pulsed electromagnetic field Lavagnino, M., Wall, M. E., Little, D., Banes, A. J., Guilak, F., & Arnoczky, S. P.
affects proliferation, tissue-Specific gene expression, and cytokines (2015). Tendon mechanobiology: Current knowledge and future
release of human tendon cells’. Cell Biochemistry and Biophysics, 66(3), research opportunities. Journal of Orthopaedic Research, 33(6),
697–708. https://doi.org/10.1007/s12013-013-9514-y 813–822. https://doi.org/10.1002/jor.22871
de Girolamo, L., Viganò, M., Galliera, E., Stanco, D., Setti, S., Marazzi, M. G., Lee, E. W. C., Maffulli, N., Li, C. K., & Chan, K. M. (1997). Pulsed magnetic
. . . Sansone, V. (2014). In vitro functional response of human tendon and electromagnetic fields in experimental achilles tendonitis in the rat:
cells to different dosages of low-frequency pulsed electromagnetic A prospective randomized study. Archives of Physical Medicine and
field’. Knee Surgery, Sports Traumatology, Arthroscopy, 23(11), Rehabilitation, 78(4), 399–404. https://doi.org/10.1016/S0003-9993
3443–3453. https://doi.org/10.1007/s00167-014-3143-x (97)90232-X
Denaro, V., Ruzzini, L., Barnaba, S. A., Longo, U. G., Campi, S., Maffulli, N., & Li, Y., Huang, G., Zhang, X., Li, B., Chen, Y., Lu, T., . . . Xu, F. (2013). Magnetic
Sgambato, A. (2011). Effect of pulsed electromagnetic fields on human hydrogels and their potential biomedical applications’. Advanced
tenocyte cultures from supraspinatus and quadriceps tendons. Ameri- Functional Materials, 23(6), 660–672. https://doi.org/10.1002/
can Journal of Physical Medicine & Rehabilitation/Association of Academic adfm.201201708
Physiatrists, 90(2), 119–127. https://doi.org/10.1097/ Li, Y., Ramcharan, M., Zhou, Z., Leong, D. J., Akinbiyi, T., Majeska, R. J., &
PHM.0b013e3181fc7bc7 Sun, H. B. (2015). The role of scleraxis in fate determination of
mesenchymal stem cells for tenocyte differentiation. Scientific Reports,
10 | PESQUEIRA ET AL.

Nature Publishing Group 5, 13149. https://doi.org/10.1038/ Randelli, P., Menon, A., Ragone, V., Creo, P., Montrasio, U. A., Orfei, C. P., . . .
srep13149 Anastasia, L. (2016). Effects of the pulsed electromagnetic field PST ®
Liu, M., Lee, C., Laron, D., Zhang, N., Waldorff, E. I., Ryaby, J. T., . . . Liu, X. on human tendon stem cells: A controlled laboratory study. BMC
(2016). Role of pulsed electromagnetic fields (PEMF) on tenocytes and Complementary and Alternative Medicine, 1–11. https://doi.org/
myoblasts-Potential application for treating rotator cuff tears. Journal of 10.1186/s12906-016- 1261-3
Orthopaedic Research, 1–21. https://doi.org/10.1002/jor.23278 Robotti, E., Zimbler, A. G., Kenna, D., & Grossman, J. A. I. (1999). The effect
Lomas, A. J., Ryan, C. N. M., Sorushanova, A., Shologu, N., Sideri, A. I., Tsioli, of pulsed electromagnetic fields on flexor tendon healing in chickens.
V., . . . Zeugolis, D. I. (2015). The past, present and future in scaffold- Journal of Hand Surgery, 24(1), 56–58.
based tendon treatments. Advanced Drug Delivery Reviews, Elsevier B.V. Ross, C. L., & Harrison, B. S. (2013). Effect of time-varied magnetic field on
84, 257–277. https://doi.org/10.1016/j.addr.2014.11.022 inflammatory response in macrophage cell line RAW 264.7. Electro-
Maeda, E., Hagiwara, Y., Wang, J. H. C., & Ohashi, T. (2013). A new magnetic Biology and Medicine, 32(1), 59–69. https://doi.org/10.3109/
experimental system for simultaneous application of cyclic tensile strain 15368378.2012.701191
and fluid shear stress to tenocytes in vitro. Biomedical Microdevices, Santos, L. J., Reis, R. L., & Gomes, M. E. (2015). Harnessing magnetic-
15(6), 1067–1075. https://doi.org/10.1007/s10544-013-9798-0 mechano actuation in regenerative medicine and tissue engineering.
Maeda, T., Sakabe, T., Sunaga, A., Sakai, K., Rivera, A. L., Keene, D. R., . . . Trends in Biotechnology, 33(8), 471–479. https://doi.org/10.1016/j.
Sakai, T. (2011). Conversion of mechanical force into TGF-beta- tibtech.2015.06.006
mediated biochemical signals. Current Biology, Elsevier Ltd 21(11), Santos, L., Silva, M., Gonçalves, A. I., Pesqueira, T., Rodrigues, M. T., &
933–941. https://doi.org/10.1016/j.cub.2011.04.007 Gomes, M. E. (2016). In vitro and in vivo assessment of magnetically
Maganaris, C. N., & Paul, J. P. (1999). In vivo human tendon mechanical actuated biomaterials and prospects in tendon healing. Nanomedicine
properties. The Journal of Physiology, 521(1), 307–313. https://doi.org/ (London, England), 11(9), 1107–1122.
10.1111/j.1469-7793.1999.00307.x Seeliger, C., Falldorf, K., Sachtleben, J., & van Griensven, M. (2014). Low-
Markov, M. S. (2007). Expanding use of pulsed electromagnetic field frequency pulsed electromagnetic fields significantly improve time of
therapies.’. Electromagnetic Biology and Medicine, 26(3), 257–274. closure and proliferation of human tendon fibroblasts. European
https://doi.org/10.1080/15368370701580806 Journal of Medical Research, 19, 37. https://doi.org/10.1186/2047-
Milet, C., & Duprez, D. (2015). The Mkx homeoprotein promotes 783X-19-37
tenogenesis in stem cells and improves tendon repair. Annals of Selvam, R., Ganesan, K., Narayana Raju, K. V. S., Gangadharan, A. C.,
Translational Medicine, 3(2), 4–7. https://doi.org/10.3978/j.issn.2305- Manohar, B. M., & Puvanakrishnan, R. (2007). Low frequency and low
5839.2015.03.64 intensity pulsed electromagnetic field exerts its antiinflammatory effect
Milovanovich, I. D., Ćirković, S., De Luka, S. R., Djordjevich, D. M., Ilić, A., through restoration of plasma membrane calcium ATPase activity. Life
Popović, T., . . . Trbovich, A. M. (2016). Homogeneous static magnetic Sciences, 80(26), 2403–2410. https://doi.org/10.1016/j.
field of different orientation induces biological changes in subacutely lfs.2007.03.019
exposed mice. Environmental Science and Pollution Research, 23(2), Silva, E. D., Babo, P. S., Costa-Almeida, R., Domingues, R. M. A., Mendes,
1584–1597. https://doi.org/10.1007/s11356-015-5109-z B. B., Paz, E., . . . Gomes, M. E. (2017). Multifunctional magnetic-
Myer, C., & Fowler, J. R. (2016). Flexor tendon repair. Healing, responsive hydrogels to engineer tendon-to-bone interface. Nano-
biomechanics, and suture configurations. Orthopedic Clinics of North medicine: Nanotechnology, Biology and Medicine, Elsevier Inc. 1–11.
America, Elsevier Inc 47(1), 219–226. https://doi.org/10.1016/j. https://doi.org/10.1016/j.nano.2017.06.002
ocl.2015.08.019 Singh, R. K., Patel, K. D., Lee, J. H., Lee, E. J., Kim, J. H., Kim, T. H., & Kim,
Osti, L., Buono, A. D., & Maffulli, N. (2015). Pulsed electromagnetic fields H. W. (2014). Potential of magnetic nanofiber scaffolds with mechanical
after rotator cuff repair: A randomized, controlled study. Orthopedics, and biological properties applicable for bone regeneration. PLoS ONE,
38(3), e223–e228. https://doi.org/10.3928/01477447-20150305-61 9(4), https://doi.org/10.1371/journal.pone.0091584
Pall, M. L. (2013). Electromagnetic fields act via activation of voltage-gated Spiesz, E. M., Thorpe, C. T., Chaudhry, S., Riley, G. P., Birch, H. L., Clegg,
calcium channels to produce beneficial or adverse effects. Journal of P. D., & Screen, H. R. C. (2015). Tendon extracellular matrix damage,
Cellular and Molecular Medicine, 17(8), 958–965. https://doi.org/ degradation and inflammation in response to in vitro overload exercise’.
10.1111/jcmm.12088 Journal of Orthopaedic Research, 33(6), 889–897. https://doi.org/
Park, J. Y., Pillinger, M. H., & Abramson, S. B. (2006). Prostaglandin E2 10.1002/jor.22879
synthesis and secretion: The role of PGE2 synthases. Clinical Immunology, Strauch, B., Patel, M. K., Rosen, D. J., Mahadevia, S., Brindzei, N., & Pilla,
119(3), 229–240. https://doi.org/10.1016/j.clim.2006.01.016 A. A. (2006). Pulsed magnetic field therapy increases tensile strength in
Pesce, M., Patruno, A., Speranza, L., & Reale, M. (2013). Extremely low a rat Achilles’ tendon repair model. The Journal of Hand Surgery, 31(7),
frequency electromagnetic field and wound healing: Implication of 1131–1135. https://doi.org/10.1016/j.jhsa.2006.03.024
cytokines as biological mediators. European Cytokine Network, 24(1), Sun, J., Kwan, R. L. C., Zheng, Y., & Cheing, G. L. Y. (2016). Effects of pulsed
1–10. https://doi.org/10.1684/ecn.2013.0332 electromagnetic fields on peripheral blood circulation in people with
Pesqueira, T., Costa-Almeida, R., & Gomes, M. E. (2017). Uncovering the diabetes: A randomized controlled trial. Bioelectromagnetics, 37(5),
effect of low-frequency static magnetic field on tendon-derived cells: 290–297. https://doi.org/10.1002/bem.21983
From mechanosensing to tenogenesis. Scientific Reports, 7(1), 10948. Suzuki, H., Ito, Y., Shinohara, M., Yamashita, S., Ichinose, S., Kishida, A., . . .
https://doi.org/10.1038/s41598-017-11253-6 Asahara, H. (2016). Gene targeting of the transcription factor Mohawk
Pilla, A. A. (2012). Electromagnetic fields instantaneously modulate nitric in rats causes heterotopic ossification of Achilles tendon via failed
oxide signaling in challenged biological systems. Biochemical and tenogenesis. Proceedings of the National Academy of Sciences of the
Biophysical Research Communications, Elsevier Inc. 426(3), 330–333. United States of America, 113(28), https://doi.org/10.1073/
https://doi.org/10.1016/j.bbrc.2012.08.078 pnas.1522054113
Popov, C., Burggraf, M., Kreja, L., Ignatius, A., Schieker, M., & Docheva, D. Tang, J. B., Zhou, Y. L., Wu, Y. F., Liu, P. Y., & Wang, X. T. (2016). Gene
(2015). Mechanical stimulation of human tendon stem/progenitor cells therapy strategies to improve strength and quality of flexor tendon
results in upregulation of matrix proteins, integrins and MMPs, and healing. Expert Opinion on Biological Therapy, 16(3), 291–301. https://
activation of p38 and ERK1/2 kinases. BMC Molecular Biology, 16(1), 6. doi.org/10.1517/14712598.2016.1134479
https://doi.org/10.1186/s12867-015- 0036-6 Tao, X., Liu, J., Chen, L., Zhou, Y., & Tang, K. (2015). EGR1 induces tenogenic
differentiation of tendon stem cells and promotes rabbit rotator cuff
PESQUEIRA ET AL.
| 11

repair. Cellular Physiology and Biochemistry, 35(2), 699–709. https://doi. Xu, D., Zhang, T., Qu, J., Hu, J., & Lu, H. (2014). Enhanced patella-patellar
org/10.1159/000369730 tendon healing using combined magnetic fields in a rabbit model. The
Thomopoulos, S., Parks, W. C., Rifkin, D. B., & Derwin, K. A. (2015). American Journal of Sports Medicine, 42(10), 2495–2501. https://doi.
Mechanisms of tendon injury and repair. Journal of Orthopaedic org/10.1177/0363546514541539
Research, 33(6), 832–839. https://doi.org/10.1002/jor.22806 Yamazaki, S., Weinhold, P. S., Graff, R. D., Tsuzaki, M., Kawakami, M.,
Thorsness, R., & Romeo, A. (2016). Massive rotator cuff tears: Trends in Minchew, J. T., & Banes, A. J. (2003). Annulus cells release ATP in
surgical management. Orthopedics, 39(3), 145–151. https://doi.org/ response to vibratory loading in vitro. Journal of Cellular Biochemistry,
10.3928/01477447-20160503-07 90(4), 812–818. https://doi.org/10.1002/jcb.10681
Tsuzaki, M., Bynum, D., Almekinders, L., Faber, J., & Banes, A. J. (2005). Yun, H.-M., Ahn, S.-J., Park, K.-R., Kim, M.-J., Kim, J.-J., Jin, G.-Z., & Kim, E.-
Mechanical loading stimulates ecto-ATPase activity in human tendon C. (2016). Magnetic nanocomposite scaffolds combined with static
cells. Journal of Cellular Biochemistry, 96(1), 117–125. https://doi.org/ magnetic field in the stimulation of osteoblastic differentiation and
10.1002/jcb.20491 bone formation. Biomaterials, Elsevier Ltd 85, 88–98. https://doi.org/
Vergallo, C., Dini, L., Szamosvölgyi, Z., Tenuzzo, B. A., Carata, E., Panzarini, 10.1016/j.biomaterials.2016.01.035
E., & László, J. F. (2013). In vitro analysis of the anti-inflammatory effect Zabloskii, V., Lunov, O., Kubinova, S., Polyakova, T., Sykova, E., & Dejneka,
of inhomogeneous static magnetic field-Exposure on human macro- A. (2016). Effects of high-gradient magnetic fields on living cell
phages and lymphocytes’. PLoS ONE, 8(8), https://doi.org/10.1371/ machinery. Journal of Physics D: Applied Physics, 49, 493003. https://doi.
journal.pone.0072374 org/10.1088/0022-3727/49/49/493003
Vincenzi, F., Targa, M., Corciulo, C., Gessi, S., Merighi, S., Setti, S., . . . Varani, Zhang, N., Lock, J., Sallee, A., & Liu, H. (2015). Magnetic nanocomposite
K. (2013). Pulsed electromagnetic fields increased the anti-inflamma- hydrogel for potential cartilage tissue engineering: synthesis, charac-
tory effect of A2A and A3 adenosine receptors in human T/C-28a2 terization, and cytocompatibility with bone marrow derived mesenchy-
chondrocytes and hFOB 1.19 osteoblasts’. PLoS ONE, 8(5), https://doi. mal stem cells. ACS Applied Materials and Interfaces, 7(37),
org/10.1371/journal.pone.0065561 20987–20998. https://doi.org/10.1021/acsami.5b06939
Wall, M. E., & Banes, A. J. (2005). Early responses to mechanical load in
tendon: Role for calcium signaling, gap junctions and intercellular
communication. Journal of Musculoskeletal Neuronal Interactions, 5(1),
How to cite this article: Pesqueira T, Costa-Almeida R,
70–84.
Xu, B., Song, G., Ju, Y., Li, X., Song, Y., & Watanabe, S. (2012). RhoA/ROCK, Gomes ME. Magnetotherapy: The quest for tendon
cytoskeletal dynamics, and focal adhesion kinase are required for regeneration. J Cell Physiol. 2018;1–11.
mechanical stretch-induced tenogenic differentiation of human mes- https://doi.org/10.1002/jcp.26637
enchymal stem cells. Journal of Cellular Physiology, 227(6), 2722–2729.
https://doi.org/10.1002/jcp.23016

Das könnte Ihnen auch gefallen