You are on page 1of 9

Available online at www.sciencedirect.

com

Status and challenges of electrical stimulation use in


chronic wound healing
Miruna Verdes 1,2, Kimberly Mace 3, Lee Margetts 4 and ]]
]]]]]]
]]

Sarah Cartmell 1,2

Non-healing wounds have led to a soaring clinical and Introduction


socioeconomical need for advanced wound-care techniques. Skin is the largest human organ. Its main function is to
Electrical stimulation is an emerging therapy inspired by the help our bodies maintain homeostasis by acting as a
wound's endogenous electric field. Promising results of clinical selective barrier between the inside and the outside,
trials have encouraged efforts to create wearable stimulation offering protection from pathogens, and damaging phy­
devices, uncover multiple cellular targets, and optimize sical and chemical agents, while also regulating the
stimulation regimes. However, the field faces a translational movement of heat, water, and electrolytes across it. This
bottleneck. This review aims to highlight the gaps between in filter function is conveyed by a complex layered struc­
vivo treatments and in vitro associated experiments by ture of interdependent cells and the matrix surrounding
discussing the current knowledge of the generation, them, with the highest adaptability and turnover being
characterization, and targets of electrical stimuli. It becomes characteristic of the two outermost layers, epidermis and
clear that enabling the translation of this technology will require dermis [1]. These are two contrasting layers, connected
increasing the complexity of the current models for skin through a highly specialized extracellular matrix (ECM)
endogenous and controlled ion transport, and investigating called the basement membrane. The epidermis relies on
which stimulus has an optimum effect on cells derived from the dermis for both mechanical stability and nutrition.
chronic wound-prone patients. Both are enhanced by the interdigitating pattern of
epidermal downgrowths (rete ridges) within the upper
Addresses dermis and dermal protrusions (papillae) within the
1
Department of Materials, Faculty of Science and Engineering, The epidermis, which give an overall increased junction
University of Manchester, Manchester M13 9PL, UK
2 surface [2]. While the structure of the epidermis mostly
The Henry Royce Institute, Royce Hub Building, The University of
Manchester, Manchester M13 9PL, UK consists of tightly interconnected cells, the dermis has a
3
Division of Cell Matrix Biology and Regenerative Medicine, Faculty of highly fibrillary architecture, within which cells are
Biology, Medicine and Health, University of Manchester, Manchester sparsely distributed [3]. Concurrently, blood vessels are
M13 9PT, UK contained within the dermis, and nutrients must diffuse
4
Department of Mechanical, Aerospace and Civil Engineering, Faculty
of Science and Engineering, The University of Manchester, Manchester
from the dermal capillary networks, through nanometer-
M13 9PL, UK sized pores in the basement membrane, to the epi­
dermis [2].
Corresponding author:
Sarah Cartmell (sarah.cartmell@manchester.ac.uk). Healthy skin, as do all the organs in our body, maintains
an electric potential difference across its epithelium.
Current Opinion in Biotechnology 2022, 75:102710 Epithelial cells in the skin, called keratinocytes, have a
This review comes from a themed issue on Tissue, cell and
heterogeneous distribution of ion channels. Ions are ac­
pathway engineering tively transported across the cell membrane against their
Edited by Lorenzo Moroni and Martijn van Griensven
electrochemical gradient to generate and preserve a
transepidermal potential ranging from 15 to 60 mV [4],
For complete overview of the section, please refer to the article
with a positive charge inside (Figure 1a). Concomitantly,
collection, “Tissue, Cell and Pathway Engineering”
some of the positively charged ions flow back to the
Available online 7th April 2022 apical side of the epithelium through the paracellular
https://doi.org/10.1016/j.copbio.2022.102710 pathway to re-establish the electrochemical equilibrium.
Crown Copyright © 2022 Published by Elsevier Ltd. All rights However, this is limited by the tight cellular junctions,
reserved. which translate into a high electrical resistivity [4].
Hence, while the epidermis is intact, the transepidermal
potential is maintained [4,5]. It has been suggested that
sodium and potassium are the main contributors to this
circuit [4], but this information is yet to be confirmed in
the skin. Conversely, measurements with ion-selective
probes conducted in corneal epithelium wounds showed

www.sciencedirect.com Current Opinion in Biotechnology 75(2022) 102710


2 Tissue, cell and pathway engineering

Figure 1

Current Opinion in Biotechnology

The skin electric field. (a) Endogenous electric potential in the intact (left) and wounded (right) skin. What we know is that keratinocytes pump ions to
maintain a transepidermal voltage gradient of 20–50 mV. Wounds are low-resistance paths through which the positive ions accumulated underneath
the epidermis can escape to the surface and generate a lateral EF. What we do not know is whether there is also a voltage gradient across the dermis
(1), and therefore do not know the input signal affecting fibroblasts, macrophages, and endothelial cells. As the layer of keratinocytes actively pumps
ions, the dermoepidermal junction could bear a positive charge relative to the deep reticular dermis. If this is true, in full-thickness wounds, there
should also be an inward positive ion flow, from the dermoepidermal junction towards the hypodermis. The newly available positive ions in the deeper
dermis (2) can now flow along the collagen fibers, and with a higher speed along the coaxial fibers in the reticular dermis, making the wound bed the
‘anode of the wound’ in comparison to the surrounding dermis. This charge transport path could in turn naturally stimulate the electrotaxis of
fibroblasts and macrophages towards the wound bed. (b) By contrast, we look at the effects of an exogenous stimulus, such as the one generated by
a wound dressing, with a central cathode and the anode on the surrounding intact skin. We predict that placing the cathode on the wound bed of a full-
thickness wound could cancel out the transepidermal potential forcing keratinocytes to pump faster (3), and reverse the ion flows (and implicitly the
cellular stimulus) both on the skin surface (4) and within the dermis (5). Here, charge density is a measure of the number of positive charges within a
unit volume.

Current Opinion in Biotechnology 75(2022) 102710 www.sciencedirect.com


Reviewing chronic wound bioelectricity principles Verdes et al. 3

that chloride, calcium, and potassium ions are actually flow into the wound, inhibiting biofilm formation, and
the main contributors to the naturally occurring outward finally directly targeting the cells to encourage a pro-
current [6]. healing behavior [21]. To date, ES has been shown to
promote angiogenesis [27–29], disrupt biofilms [30], and
Damage to the skin is not uncommon, and complex induce various responses in the cells involved in wound
processes ensure that the skin barrier is restored in a healing (Tables 1 and 2). These results should en­
timely fashion. Our cells have evolved to recognize the courage further exploration, as different stimulation
breach and synchronize with their neighbors to stop the techniques may lead to better results. Here, the pitfalls
bleeding, clean up damaged tissue, and generate new and challenges of using ES to target the different cellular
dermal ECM on top of which keratinocytes can migrate agents of wound healing will be discussed.
and close the wound before pathogens can become es­
tablished. After re-epithelialization, in the absence of
imminent pathogen threats, the mechanical properties of The signal
the skin are restored through a long process of re­ The EF and the resulting endogenous ionic currents are
modeling [7,8]. one of multiple wound-healing guiding cues. Changes in
the local skin EF appear immediately upon wounding,
However, this process can often fail, and non-healing and are gradually dampened as the wound heals and the
wounds are a growing societal burden. In the UK alone, epidermal resistance is recovered [4]. Cells that play a
the annual prevalence of wounds increased by 71% be­ key role in wound healing, such as keratinocytes [31],
tween 2012 and 2013 and 2017 and 2018, and 67% of the macrophages [32], fibroblasts [33], and endothelial cells
£8.3 billion budget allocated for wound care in the latter [34,35], were shown to react to such changes.
period was spent on wounds that did not heal within that
year [9]. Several intrinsic factors make the skin less re­ Aiming to reproduce the healing benefits of the en­
sistant to mechanical stress, and impede healing, as most dogenous EF, we developed a range of ES devices for
chronic wounds are either pressure, diabetic, or venous the in vivo stimulation of chronic wounds [26]. The ad­
skin ulcers [10]. These comorbidities may lead to a vantage of these over other guiding cues, such as damage
suboptimal ECM structure [11,12] and cellular behavior or pathogen-associated molecular patterns, or various cell
[13–20], which can then contribute to thinning of the signaling molecules is that exogenous ES can in theory
epidermis, reduced skin elasticity, tissue necrosis, and be more precisely controlled. The current ES devices
eventually wounds [11]. In addition, when the epidermal consist of electrodes in direct contact with the skin
barrier is breached for a long time, wound beds are [26,36], with the option of adding state-of-the-art tech­
further disturbed by bacterial colonies forming biofilms nology such as nanogenerators, nanostructured con­
[21]. The wound then fails to exit the inflammatory ductive dressings, or nanoparticles with ES-dependent
phase, whereby the ECM is excessively degraded, and drug release to improve the wearability, compatibility,
the wound cells become senescent and less responsive to and functionality [37,38]. The cathode is generally
stimuli [7,22]. placed in the middle of the wound, while the anode
either surrounds the wound edges or is placed on one
Acute skin wounds generate a lateral electric field (EF) side of the wound [26].
of about 100–200 V/m, with the wound edge being more
positive than the center [4,23], as ions escape through Although any electric potential gradient can be gener­
the epidermal breach and higher current densities are ated with adequate electric devices, we are still limited
recorded on the wound outline (Figure 1a). This, cou­ in controlling the cellular input in vivo by the scarce
pled with the observed keratinocyte migration towards knowledge regarding the electrical properties of the
the cathode of an EF, encouraged the development of transduction path. Firstly, even though the electrical
electroactive wound dressings in which the cathode is properties of the skin are sensitive to physiological
connected to the wound bed and the anode to the edges changes and vary significantly between the body loca­
and surrounding skin (Figure 1b). Measurements in tions of the same individual [39,40], to date there is no
corneal wounds of diabetic mice suggest that the EF is study that has measured the electrical properties in pa­
indeed weaker in some chronic wounds [24]. However, tients with chronic ulcers, or in locations prone to such
measurements of the endogenous EF in chronic wounds wounds (sacral region or legs). Secondly, the only in vivo
remain a fundamental gap in the literature. skin EF sensors, the vibrating probe [41] and the Der­
macorder [23], can only measure the EF at the wound
Although electrical stimulation (ES) is a versatile tool for surface. While these are useful in determining the EF
wound healing, devices and treatment regimens still impacting the keratinocytes at the wound edge and
need to be optimized for this therapy to reach its full bacteria settling on the wound surface, they do not
potential [25,26]. An effective chronic wound treatment provide information about the EF of the deeper dermis.
should target ischemia by enhancing nutrient-rich blood This information is important, as current electroceuticals

www.sciencedirect.com Current Opinion in Biotechnology 75(2022) 102710


4 Tissue, cell and pathway engineering

may in fact dampen or even reverse the directional


Reference
electric cue arriving at fibroblasts and macrophages re­
[64] sident in the dermis surrounding the wound (Figure 1b).

[67]

[66]

[27]

[65]

[33]

[70]
constant, polymerized actin, and podosomes polarized toward the
In silico models can predict the endogenous EF [42] or

Anode (preceded by elongated morphology perpendicular to EF)


Anode (preceded by elongated morphology with total cell area

the impact of electrodes applied to wounded skin within


the deeper layers [43,44]. However, there is still space
for improvement, as these models do not account for the
Perpendicular to EF (laminin and fibronectin substrate)

ability of keratinocytes to maintain the transepidermal


potential gradient, a physiological charge distribution
within the dermis, and assume that the skin layers are
isotropic materials when this may not be the case [45]. In
silico simulations of charge transport combined with
Anode (peaking at 4 hours of ES)
high-precision ECM images [46] should now constitute a
viable solution for building a more representative model
leading edge, facing the anode)

of ion conduction within and around wounds. Validation


Cathode (Ca2+ independent)
Random (glass substrate)

of these models may be done using the methods for


Avg speed: 0.05 µm/min

Avg speed: 0.15 µm/min

Enhanced (Scratch test)


studying the endogenous ion distribution within the
Direction of migration

Random enhanced

skin [47].
DC 1 hour, 100 V/m Not significant

Various in vitro cellular ES techniques have also been


DC, 2 hours 150 V/m Cathode

developed. Classing by the EF generation method,


DC 1 hour, 400 V/m Anode

these involve either direct, capacitive, inductive cou­


pling, or stimulation via conductive scaffolds [36,48,49].
While in vitro setups offer better control over the ex­
DC up to 6 hours,
AC, 1 Hz, 90 min,

perimental conditions, the complexity of the cell culture


medium still makes the characterization of the cellular
DC, 24 hours,

DC > 1 hour,
100–400 V/m

150–200 V/m

25–100 V/m
DC, 10 min,
Electrotaxis of monocytes, macrophages and fibroblasts in response to electrical stimulation.

DC, 1 hour,

electric microenvironment challenging. Therefore, it is


200 V/m

100 V/m

200 V/m

still difficult to compare the results from different in vitro


methods on one hand, and unclear how well they model
existent electroceuticals’ effect on the skin in vivo on the
other hand. In silico models can also assist the char­
Ag/AgCl electrodes and calf

Conductive PPy/HE/PLLA

acterization of cellular electric microenvironments in


Stimulus

vitro, but their limitations must not be forgotten.


Platinum electrodes
Direct coupling Ag/AgCl electrodes

Ag/AgCl electrodes

collagen I coating

For the direct coupling setup, a common simple ap­


proximation of the stimulus is the ratio between the
membranes

voltage gradient applied on the electrodes and the dis­


tance between them. At the same time, the generated
stimulus can also be spatially resolved at different levels
of precision using commercially available software
packages that are used to model electrolytic cells
ES strategy

ES through

[50–52]. Contrary to what the first approximation im­


scaffold

plies, the in silico simulations show that the generated


EF is not homogenous, leading to higher current den­
sities near the electrodes [50,51]. In vitro, this translates
Bovine pulmonary artery
Fibroblasts Murine NIH 3T3&SV101

into cell death or different cellular responses throughout


the culture dish, showing the need to delimit the culture
Human dermal

area [50]. In vivo, this should argue for developing me­


(embryonic)

chanisms that can ensure that currents in all wound areas


Human macrophages

are maintained underneath a biocompatible threshold.


Human monocyte

While current in silico models have proved useful in this


case, they are still an approximation, as the culture
medium is a concentrated electrolyte with multiple
Table 1

charge carriers and macromolecules that may impact the


Cell

transduction of time-dependent ES.

Current Opinion in Biotechnology 75(2022) 102710 www.sciencedirect.com


Reviewing chronic wound bioelectricity principles Verdes et al. 5

Table 2
Functionality of macrophages and fibroblasts harnessed through electrical stimulation.

Cell ES strategy Stimulus Cell response Ref


Human Direct coupling with No extra stimulus DC, 2 hours, 150 V/m Phagocytosis enhanced [64]
macrophage Ag/AgCl electrodes After 24 hours:
TNF-α: moderate increase (M1-like)
NT3: significant increase (M2-like)
CCL2, IL10, VEGF, TGF-β: constant
PI3K and p-ERK activated
IFN-γ and LPS (M1 IL1b increased
stimuli) IL-23 increased
IL-6 constant
IL-10 constant
TNF-a constant
IL-4 (M2 stimulus) TNF-α enhanced
VEGF constant
IL-10 constant
TGF-b constant
Human dermal PCL scaffold with EF Step pulses, 3 hours, α-SMA 12.4× higher expression [71]
fibroblast aligned uniaxial fibers 50 V/m FGF2, EGF, FN secretion elevated
and TGF-β1 (compared with just TGF-β1 and scaffold)
ES through scaffold Conductive PPy/HE/ DC, 6 hours, 200 V/m Contractility enhanced [70]
PLLA membranes α-SMA expression enhanced (starting at
2 hours ES)
FGF1, FGF2 secretion enhanced
DC, 6 hours, Production increased: CCL7, KGF, TIMP2 [69]
50–200 V/m Production decreased: MMP2
Ppy-coated fabrics Step pulses, 24 hours, TGF-β1, p-ERK, NF-kB upregulated [72]
100 V/m Changes maintained in subculture and in
vivo transplants
Direct coupling Carbon rod electrodes Step pulses, Collagen, MMP1, elastin significant [73]
Confluence ≥ 90% 0.5–24 hours, expression increase at applied EF above
0–1000 V/m 300 V/m, and stimulation longer than
2 hours

In other in vitro models, electrolysis is avoided to pre­ are highly proliferative at the wound edges, they fail to
vent generating harmful byproducts [49], but stimulus migrate over the wound bed and seal the lesion [56].
characterization becomes more challenging. Briefly, cell While ES is supposed to override this problem, in vitro,
culture wells can be placed between the plates of a ca­ keratinocyte electrotaxis is blocked by the absence of
pacitor (capacitive coupling), near an inductor coil (in­ extracellular calcium [57], and, additionally, the direc­
ductive coupling), or on top of a biocompatible tion can be reversed by lowering the extracellular pH
conductive scaffold. While the interaction of capacitor [58]. Conversely, as the wound heals, the pH moves
plates with dielectric materials and inductor coils with from neutral to acidic, and wounds with a higher pH are
conductors is well understood, new models are required shown to heal slower [59,60]. Moreover, even if the
to describe their interaction with electrolytes within the surface pH is right, keratinocyte electrotaxis may still be
cell culture recipients. Similarly, the principles of in­ inhibited by inflammatory cytokines in the wound bed,
teraction of conductive polymers with biological cells or, in full-thickness wounds where the dermal layer is
and their electrolytic culture medium are yet to be de­ damaged, by an incomplete granulation tissue that they
scribed and modeled. As such, even though electricity is cannot connect with [61]. These are both problems that
involved in all those in vitro setups, it is not clear whe­ could be introduced by suboptimal macrophage and fi­
ther they all generate the same cellular stimulus, how to broblast behavior in chronic wounds.
compare them, or how well they are translated into
current electroceuticals. Successful wound healing and re-epithelialization are
dependent on the dynamic evolution of the in­
The targets flammatory phase and transition to the repair phase, a
The first studies to suggest that ES can promote wound process tightly regulated by macrophages and fibroblasts
healing were focused on epithelial cells, such as kerati­ [8,62]. The former is characterized by the majority of
nocytes in the skin [4]. Several studies have shown that macrophages exhibiting a pro-inflammatory phenotype
keratinocytes migrate toward the cathode of an EF that promotes host defense and removal of damaged
[53–55]. In chronic wounds, even though keratinocytes tissue while fibroblasts are recruited to the provisional

www.sciencedirect.com Current Opinion in Biotechnology 75(2022) 102710


6 Tissue, cell and pathway engineering

Table 3
Cellular electrical properties and their fast response to ES.

Cell Device Stimulus Cell response Reference


Macrophage Human Glass None Resting membrane potential [−49, −72] mV [83]
microelectrode Depolarizing current Repetitive Ca2+-dependent spikes [83,84]
12.7–30.5 V/s
Direct coupling DC, 2 hours 150 V/m Gradual rise in intracellular Ca2+ peaking at 2.4 min [64]
Ag/AgCl electrodes of ES
Murine Patch clamp Voltage pulses and ramp A computational model for the macrophage's [82]
cardiac −100 to +60 mV electrophysiological properties was developed.
Reported properties are:
▪ membrane resistance 2.2 ± 0.1 GΩ
▪ capacitance 18.3 ± 0.1 pF
▪ resting membrane potential − 39.6 ± 0.3 mV
Human fibroblast Direct coupling 1 Hz; 30 min, 200 V/m Integrin redistributed around the nucleus [32]
Platinum electrodes DC; short term, 1000 V/m Ca2+ inflow (probably through VGCC)
AC; 1 Hz; short term,
1000 V/m
AC; 1 Hz; 30 min, 200 V/m
AC; 10 Hz; 30 min,
200 V/m
AC; 100 Hz; 30 min, No significant increase in intracellular Ca2+
200 V/m

ECM wound bed. Subsequently, within the latter phase, Functionality has also been regulated with ES in vitro
the pro-repair macrophages become the predominant (Table 2). Both macrophages and fibroblasts are highly
phenotype. At the same time, the recruited fibroblasts plastic cells that change their phenotype and associated
degrade the provisional ECM fibrin, replace it with function as the healing signs of progress [62,68]. Direct
collagen-rich granulation tissue, convert it to myofibro­ current or low frequency ES triggers Ca2+ intake
blasts, and contract the wound [63]. Both macrophages (Table 3), and impacts cytokine, enzyme, and matrix
and fibroblasts secrete several other growth factors, protein secretion in both cell types [35,64,69–73]. At the
which can, in turn, regulate re-epithelialization and an­ same time, macrophage phagocytosis is more efficient
giogenesis by their impact on keratinocytes and en­ [64], and the contractility of fibroblasts is increased as
dothelial cells. Therefore, only these two will be further they differentiate into myofibroblasts [70,71,74]. Subse­
focused on as ES targets in chronic wound healing. quently, myofibroblasts could promote the transition to
the macrophage pro-repair phenotype [62,75,76].
In vitro experiments show that recruitment of both
macrophages and fibroblasts to the wound bed could be However, while most ES devices aim to recreate the
targeted with direct current (DC) ES of physiological endogenous EF recorded in the wounds of healthy skin,
strength (Table 1). However, they require a longer and it is unlikely that cells in chronic wounds will react the
opposite stimulus to keratinocytes, a fact that might not same as their healthy counterparts [13–18]. Both mac­
be addressed by some in vivo treatments and wound rophages and fibroblasts undergo profound changes in
dressings (Figure 1b). Both macrophages and fibroblasts patients prone to chronic wounds [13–18], but their ef­
exhibit electrotaxis toward the anode of a DC EF in a fects on the electrical properties have not been de­
voltage-dependent manner [33,64], and require more scribed yet. Membrane-bound voltage-sensitive dyes
than an hour of ES to begin migrating directionally [77–79], ion-bound fluorescent dyes [32,64,66], ion
[33,64,65]. Moreover, establishing the origin and differ­ channel blockers [80], microfluidic devices [81], and
entiation status of the targeted cells will play an im­ patch-clamp electrodes [82–84] could be used to quan­
portant role in the in vivo chronic wound therapy results. tify those changes in single cells. While current data
In vitro, while mature macrophages migrate toward the show the pro-healing effects of ES on healthy cells, the
anode [64], their bone marrow-derived progenitors, stimulus must be optimized to trigger responses in cells
monocytes, are directed toward the cathode [64]. Simi­ derived from chronic wound-prone patients or model
larly, fibroblasts from embryonic-origin cell lines were organisms.
also reported to migrate toward the cathode [66]. On top
of this, the way in which the tissue transduces the exo­ Conclusion
genous stimulus may also impact the recruitment result. In this short review, the limitations and opportunities in
Mature macrophages exposed to an alternating current the characterization of the electrical stimulus for wound-
EF migrate perpendicular to the field direction [67]. healing therapies have been outlined. Moreover, it is

Current Opinion in Biotechnology 75(2022) 102710 www.sciencedirect.com


Reviewing chronic wound bioelectricity principles Verdes et al. 7

argued that for chronic wounds, especially the full- 6. Vieira AC, Reid B, Cao L, Mannis MJ, Schwab IR, Zhao M: Ionic
thickness ones where the dermal layer is damaged and components of electric current at rat corneal wounds. PLOS
One 2011, 6:e17411.
inflammatory processes are dysregulated, it is not suffi­
7. Eming SA, Martin P, Tomic-Canic M: Wound repair and
cient to only estimate the stimulus influencing the regeneration: mechanisms, signaling, and translation. Sci
wound-edge keratinocytes, and thus only have a control Transl Med 2014, 6:265sr6.
mechanism for the epithelial layer. Considering this, a 8. Cañedo-Dorantes L, Cañedo-Ayala M: Skin acute wound healing:
natural step in advancing wound ES treatment protocols a comprehensive review. Int J Inflam 2019, 2019:3706315.
is targeting the cells that orchestrate dermal repair, 9. Guest JF, Fuller GW, Vowden P: Cohort study evaluating the
namely macrophages and fibroblasts. While both these burden of wounds to the UK’s National Health Service in 2017/
2018: update from 2012/2013. BMJ Open 2020, 10:1-15.
cell types were shown to react to an applied EF, suc­
cessful optimization of devices and treatment protocols 10. Eaglstein WH, Falanga V: Chronic wounds. Surg Clin North Am
1997, 77:689-700.
still requires interdisciplinary effort in two major bodies
11. Chao CYL, Zheng YP, Cheing GLY: Epidermal thickness and
of work. The first one is in modeling the transduction of biomechanical properties of plantar tissues in diabetic foot.
the electric stimulus, from the electrodes to the target Ultrasound Med Biol 2011, 37:1029-1038.
cells, both in vitro and in vivo. Crucially here, better 12. Pai S, Ledoux WR: The compressive mechanical properties of
characterization of the dermal EF must be performed in diabetic and non-diabetic plantar soft tissue. J Biomech 2010,
43:1754-1760.
order to understand the endogenous microenvironment
in which the macrophages and fibroblasts reside. The 13. Nikiforov NG, Galstyan KO, Nedosugova LV, Elizova NV,
Kolmychkova KI, Ivanova EA: Proinflammatory monocyte
second one is in characterizing the ES response of those polarization in type 2 diabetes mellitus and coronary heart
cells derived from patients prone to chronic wounds. disease. Vessel Plus 2017, 1:192-195, https://doi.org/10.20517/
2574-1209.2017.21
Thankfully, a range of tools is already available.
Computational models, enriched with microscopy-en­ 14. Pang J, Maienschein-Cline M, Koh TJ: Enhanced proliferation of
Ly6C + monocytes/macrophages contributes to chronic
abled detailed geometry, can guide ES application in inflammation in skin wounds of diabetic mice. J Immunol 2021,
vitro, help launch hypotheses of ionic flow within the 206:621-630.
skin, and help estimate the cellular electrical properties, 15. Torbica T, Wicks K, Umehara T, Gungordu L, Alrdahe S, Wemyss K,
as well as their changes because of aging and chronic Grainger JR, Mace KA: Chronic inflammation in response to
injury: retention of myeloid cells in injured tissue is driven by
diseases. Validation can be sought with ion distribution myeloid cell intrinsic factors. J Invest Dermatol 2019,
experiments in vitro at a single-cell level, or in ex vivo 139:1583-1592.
skin. Eventually, these efforts will inform and facilitate 16. Qing C: The molecular biology in wound healing & non-healing
the design of a new generation of electroceuticals to treat wound. Chinese J Traumatol — English Ed 2017, 20:189-193.
chronic wounds. 17. Lerman OZ, Galiano RD, Armour M, Levine JP, Gurtner GC:
Cellular dysfunction in the diabetic fibroblast: impairment in
migration, vascular endothelial growth factor production, and
Conflict of interest statement response to hypoxia. Am J Pathol 2003, 162:303-312.
None declared. 18. Berberich B, Thriene K, Gretzmeier C, Kü Hl T, Bayer H, Athanasiou
I, Rafei-Shamsabadi DA, Bruckner-Tuderman L, Nyströ A, Kiritsi D,
Acknowledgements et al.: Proteomic profiling of fibroblasts isolated from chronic
wounds identifies disease-relevant signaling pathways. J Invest
This work was supported by the Henry Royce Institute for Advanced Dermatol (11) 2020, 140:2280-2290, https://doi.org/10.1016/j.jid.
Materials, funded through Engineering and Physical Sciences Research 2020.02.040.
Council (EPSRC) grants EP/R00661X/1, EP/P025021/1 and EP/P025498/1, •Using proteomic profiling and functional characterization, the authors
and by the 4-year Wellcome Trust PhD Programme in Quantitative & show that dermal fibroblasts isolated from human chronic wounds (re­
Biophysical Biology. sulting from trauma or venous insufficiency) exhibit a significantly dif­
ferent phenotype compared with dermal fibroblasts isolated from
healthy skin biopsies or acute wounds.
References and recommended reading 19. De Maeyer RPH, Chambers ES: The impact of ageing on
Papers of particular interest, published within the period of review, have
monocytes and macrophages. Immunol Lett 2021, 230:1-10.
been highlighted as:
20. Ong SM, Hadadi E, Dang TM, Yeap WH, Tan CTY, Ng TP, Larbi A,
• of special interest Wong SC: The pro-inflammatory phenotype of the human non-
•• of outstanding interest. classical monocyte subset is attributed to senescence article.
Cell Death Dis 2018, 9:1-12.
1. Nyström A, Bruckner-Tuderman L: Matrix molecules and skin
biology. Semin Cell Dev Biol 2019, 89:136-146. 21. Mustoe T: Understanding chronic wounds: a unifying
hypothesis on their pathogenesis and implications for therapy.
2. Roig-Rosello E, Rousselle P: The human epidermal basement Am J Surg 2004, 187:S65-S70.
membrane: a shaped and cell instructive platform that aging
slowly alters. Biomolecules 2020, 10:1-32. 22. Eming SA, Koch M, Krieger A, Brachvogel B, Kreft S, Bruckner-
Tuderman L, Krieg T, Shannon JD, Fox JW: Differential proteomic
3. Kleiser S, Nyström A: Interplay between cell-surface receptors analysis distinguishes tissue repair biomarker signatures in
and extracellular matrix in skin. Biomolecules 2020, 10:1-39. wound exudates obtained from normal healing and chronic
wounds. J Proteome Res 2010, 9:4758-4766.
4. Nuccitelli R: A role for endogenous electric fields in wound
healing. Curr Top Dev Biol 2003, 58:1-26. 23. Pullar CE: The Physiology of Bioelectricity in Development,
Tissue Regeneration and Cancer. CRC Press; 2011.
5. Nuccitelli R: Endogenous ionic currents and DC electric fields in
multicellular animal tissues. Bioelectromagnetics 1992, 24. Shen Y, Pfluger T, Ferreira F, Liang J, Navedo MF, Zeng Q, Reid B,
13:147-157. Zhao M: Diabetic cornea wounds produce significantly weaker

www.sciencedirect.com Current Opinion in Biotechnology 75(2022) 102710


8 Tissue, cell and pathway engineering

electric signals that may contribute to impaired healing. Sci impedance in regions prone to chronic wounds, is essential for further
Rep 2016, 6:1-12. ES therapeutic advances.
25. Arora M, Harvey LA, Glinsky JV, Nier L, Lavrencic L, Kifley A, 41. Reid B, Nuccitelli R, Zhao M: Non-invasive measurement of
Cameron ID: Electrical stimulation for treating pressure ulcers. bioelectric currents with a vibrating probe. Nat Protoc 2007,
Cochrane Database Syst Rev 2020,2020. 2:661-669.
26. Kloth LC: Electrical stimulation technologies for wound healing. 42. Karba R, Šemrov D, Vodovnik L, Benko H, Šavrin R: DC electrical
Adv Wound Care 2014, 3:81-90. stimulation for chronic wound healing enhancement. Part 1.
27. Bai H, McCaig CD, Forrester JV, Zhao M: DC electric fields Clinical study and determination of electrical field distribution
induce distinct preangiogenic responses in microvascular and in the numerical wound model. Bioelectrochem Bioenergy 1997,
macrovascular cells. Arterioscler Thromb Vasc Biol 2004, 43:265-270.
24:1234-1239. 43. Šemrov D, Karba R, Valenčič V: DC electrical stimulation for
28. Ud-Din S, Sebastian A, Giddings P, Colthurst J, Whiteside S, Morris chronic wound healing enhancement. Part 2. Parameter
J, Nuccitelli R, Pullar C, Baguneid M, Bayat A: Angiogenesis is determination by numerical modelling. Bioelectrochem
induced and wound size is reduced by electrical stimulation in Bioenergy 1997, 43:271-277.
an acute wound healing model in human skin. PLOS One 2015,
44. Sun YS: Electrical stimulation for wound-healing: simulation on
10:1-22.
the effect of electrode configurations. Biomed Res Int 2017,
29. Asadi MR, Torkaman G, Hedayati M, Mohajeri-Tehrani MR, Ahmadi 2017:5289041.
M, Gohardani RF: Angiogenic effects of low-intensity cathodal
direct current on ischemic diabetic foot ulcers: a randomized 45. Tavernier A, Dierickx M, Hinsenkamp M: Tensors of dielectric
controlled trial. Diabetes Res Clin Pract 2017, 127:147-155. permittivity and conductivity of in vitro human dermis and
epidermis. Bioelectrochem Bioenergy 1993, 30:65-72.
30. Ashrafi M, Novak-Frazer L, Morris J, Baguneid M, Rautemaa-
Richardson R, Bayat A: Electrical stimulation disrupts biofilms in 46. Poole JJA, Mostaço-Guidolin LB: Optical microscopy and the
a human wound model and reveals the potential for monitoring • extracellular matrix structure: a review. Cells 2021, 10:1760.
treatment response with volatile biomarkers. Wound Repair •A comprehensive review of optical microscopy techniques and how
Regen 2019, 27:5-18. they can be used to investigate the structure of the extracellular matrix.

31. Rouabhia M, Park HJ, Abedin-Do A, Douville Y, Méthot M, Zhang Z: 47. Tarnowska M, Briançon S, Resende de Azevedo J, Chevalier Y,
Electrical stimulation promotes the proliferation of human Bolzinger MA: Inorganic ions in the skin: allies or enemies? Int J
keratinocytes, increases the production of keratin 5 and 14, Pharm 2020, 591:119991.
and increases the phosphorylation of ERK1/2 and p38 MAP
kinases. J Tissue Eng Regen Med 2020, 14:909-919. 48. Chen C, Bai X, Ding Y, Lee IS: Electrical stimulation as a novel
tool for regulating cell behavior in tissue engineering. Biomater
32. Cho MR: A review of electrocoupling mechanisms mediating Res 2019, 23:25.
facilitated wound healing. IEEE Trans Plasma Sci 2002,
30:1504-1515. 49. Balint R, Cassidy NJ, Cartmell SH: Electrical stimulation: a novel
tool for tissue engineering. Tissue Eng — Part B Rev 2013,
33. Guo A, Song B, Reid B, Gu Y, Forrester JV, Jahoda CAB, Zhao M: 19:48-57.
Effects of physiological electric fields on migration of human
dermal fibroblasts. J Investig Dermatol 2010, 130:2320-2327. 50. Srirussamee K, Cassidy NJ, Cartmell SH: Direct electrical
stimulation enhances osteogenesis by inducing Bmp2 and
34. Sheikh AQ, Taghian T, Hemingway B, Cho H, Kogan AB, Spp1 expressions from macrophages and preosteoblasts.
Narmoneva DA: Regulation of endothelial MAPK/ERK signalling Biotechnol Bioeng 2019, 116:3421-3432, https://doi.org/10.1002/
and capillary morphogenesis by low-amplitude electric field. J bit.27142
R Soc Interface 2013, 10:20120548.
51. Srirussamee K, Xue R, Mobini S, Cassidy NJ, Cartmell SH:
35. Bostan LE, Almqvist S, Pullar CE: A pulsed current electric field Changes in the extracellular microenvironment and osteogenic
•• alters protein expression creating a wound healing phenotype responses of mesenchymal stem/stromal cells induced by in
in human skin cells. Regen Med 2020, 15:1611-1623. vitro direct electrical stimulation. J Tissue Eng 2021, 12:1-17,
••Using the RT2 Profiler PCR Array for Human Wound Healing and https://doi.org/10.1177/2041731420974147
specific ELISA kits, the authors report that pulsed current electrical
stimulation significantly altered the expression in 31 genes and 7 52. Verdes M, Disney C, Phamornnak C, Margetts L, Cartmell S: Finite
proangiogenic and proregenerative growth factors in response to sti­ element modelling of a cellular electric microenvironment. J Vis
mulation of keratinocytes/dermal fibroblasts/dermal microvascular en­ Exp 2021, 2021:e61928.
dothelial cells and macrophages. This is the first study that investigates
the effect of ES on such a large number of genes. 53. Nishimura KY, Isseroff RR, Nucciteili R: Human keratinocytes
migrate to the negative pole in direct current electric fields
36. Zhao S, Mehta AS, Zhao M: Biomedical applications of electrical comparable to those measured in mammalian wounds. J Cell
stimulation. Cell Mol Life Sci 2020, 77:2681-2699. Sci 1996, 109:199-207.
37. Hunckler J, de Mel A: A current affair: electrotherapy in wound
54. Ren X, Sun H, Liu J, Guo X, Huang J, Jiang X, Zhang Y, Huang Y,
healing. J Multidiscip Healthc 2017, 10:179-194. Fan D, Zhang J: Keratinocyte electrotaxis induced by
38. Luo R, Dai J, Zhang J, Li Z: Accelerated skin wound healing by physiological pulsed direct current electric fields.
electrical stimulation. Adv Healthc Mater 2021, 10:1-15. Bioelectrochemistry 2019, 127:113-124.

39. Grant JP, Clarke RN, Symm GT, Spyrou NM: In vivo dielectric 55. Zajdel TJ, Shim G, Cohen DJ: Come together: on-chip bioelectric
properties of human skin from 50 MHz to 2.0 GHz. Phys Med • wound closure. Biosens Bioelectron 2021, 192:113479.
Biol 1988, 33:607-612. •Presents a compact, low-cost bioreactor specifically designed for real-
time studies of electrotaxis in the context of wound healing. This is the
40. Naqvi SAR, Manoufali M, Mohammed B, Mobashsher AT, Foong D, first in vitro electrical stimulation device that generates a convergent
Abbosh AM: In vivo human skin dielectric properties field geometry, making it a closer approximation of the endogenous
characterization and statistical analysis at frequencies from 1 electric field.
to 30 GHz. IEEE Trans Instrum Meas 2021, 70:6001710.
••Using the coaxial probe-based reflection method, the authors mea­ 56. Martin P, Nunan R: Cellular and molecular mechanisms of repair
sured the skin impedance of 50 healthy volunteers, in several regions of in acute and chronic wound healing. Br J Dermatol 2015,
interest on their hands. On the basis of statistical analysis, the authors 173:370-378.
show that different skin models are required to accurately represent
different regions on the human body, and that in the case of hands, the 57. Fang KS, Farboud B, Nuccitelli R, Isseroff RR: Migration of human
dielectric skin properties depend on gender but not on weight and age. keratinocytes in electric fields requires growth factors and
A similar study, looking at the effects of age and diabetes on the skin extracellular calcium. J Invest Dermatol 1998, 111:751-756.

Current Opinion in Biotechnology 75(2022) 102710 www.sciencedirect.com


Reviewing chronic wound bioelectricity principles Verdes et al. 9

58. Saltukoglu D, Grünewald J, Strohmeyer N, Bensch R, Ulbrich MH, 73. Nguyen EB, Wishner J, Slowinska K: The effect of pulsed electric
Ronneberger O, Simons M: Spontaneous and electric field- field on expression of ECM proteins: collagen, elastin, and
controlled front-rear polarization of human keratinocytes. Mol MMP1 in human dermal fibroblasts. J Electroanal Chem 2018,
Biol Cell 2015, 26:4373-4386. 812:265-272.
59. Gethin G: The significance of surface pH in chronic wounds. 74. Han SJ, Moon D, Park MY, Kwon S, Noh M, Jang J, Lee JB, Kim
Wounds 2007, 3:52-56. KS: Electric field-induced changes in biomechanical properties
in human dermal fibroblasts and a human skin equivalent. Ski
60. Greener B, Hughes AA, Bannister NP, Douglass J: Proteases and Res Technol 2020, 26:914-922.
pH in chronic wounds. J Wound Care 2005, 14:59-61.
75. Xue J, Sharma V, Hsieh MH, Chawla A, Murali R, Pandol SJ,
61. Krejci-Papa NC, Hoang AN, Hansbrough JF: Fibroblast sheets Habtezion A: Alternatively activated macrophages promote
enable epithelialization of wounds that do not support pancreatic fibrosis in chronic pancreatitis. Nat Commun 2015,6.
keratinocyte migration. Tissue Eng 1999, 5:555-561.
76. Humeres C, Vivar R, Boza P, Muñoz C, Bolivar S, Anfossi R, Osorio
62. Schuster R, Rockel JS, Kapoor M, Hinz B: The inflammatory JM, Olivares-Silva F, García L, Díaz-Araya G: Cardiac fibroblast
speech of fibroblasts. Immunol Rev 2021, 302:126-146, https:// cytokine profiles induced by proinflammatory or profibrotic
doi.org/10.1111/imr.12971. stimuli promote monocyte recruitment and modulate
••An enlightening review on fibroblasts and their role in regenerative macrophage M1/M2 balance in vitro. J Mol Cell Cardiol 2016,
processes. Their multiple precursors, competing cellular states, and 101:69-80.
bidirectional interactions with macrophages, crucial to inflammation and
repair, are presented. 77. Liu P, Miller EW: Electrophysiology, unplugged: imaging
membrane potential with fluorescent indicators. Acc Chem Res
63. Singh S, Young A, McNaught CE: The physiology of wound 2020, 53:11-19.
healing. Surgery 2017, 35:473-477.
78. Lazzari-Dean JR, Gest AMM, Miller EW: Optical estimation of
64. Hoare JI, Rajnicek AM, McCaig CD, Barker RN, Wilson HM: •• absolute membrane potential using fluorescence lifetime
Electric fields are novel determinants of human macrophage imaging. Elife 2019, 8:1-29.
functions. J Leukoc Biol 2016, 99:1141-1151. ••The authors develop a novel method for estimating cellular trans­
65. Snyder S, DeJulius C, Willits RK: Electrical stimulation increases membrane potentials using fluorescence lifetime imaging of
random migration of human dermal fibroblasts. Ann Biomed VoltageFluor dyes. This is a high-throughput method that can be cali­
Eng 2017, 45:2049-2060. brated for different cell lines, to report the resting membrane potentials
of thousands of cells at single-cell resolution, with 10–23 mV accuracy.
66. Brown MJ, Loew LM: Electric field-directed fibroblast
locomotion involves cell surface molecular reorganization and 79. Lazzari-Dean JR, Miller EW: Optical estimation of absolute
is calcium independent. J Cell Biol 1994, 127:117-128. •• membrane potential using one- and two-photon fluorescence
lifetime imaging microscopy. Bioelectricity 2021, 3:197-203.
67. Cho MR, Thatte HS, Lee RC, Golan DE: Integrin-dependent ••Building on their previous work referenced above, the authors com­
human macrophage migration induced by oscillatory electrical pare the results obtained with fluorescence lifetime imaging of
stimulation. Ann Biomed Eng 2000, 28:234-243. VoltageFluor dyes on different devices, to prove its consistency across
platforms. Notably, the authors demonstrate that cellular transmem­
68. Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, brane potential can be accurately reported with two-photon illumination,
Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, setting the foundation for this method’s use in thick tissue.
Sahebkar A: Macrophage plasticity, polarization, and function in
health and disease. J Cell Physiol 2018, 233:6425-6440. 80. Li C, Levin M, Kaplan DL: Bioelectric modulation of macrophage
polarization. Sci Rep 2016, 6:1-12.
69. Park HJ, Rouabhia M, Lavertu D, Zhang Z: Electrical stimulation
modulates the expression of multiple wound healing genes in 81. Spencer D, Morgan H: High-speed single-cell dielectric
primary human dermal fibroblasts. Tissue Eng — Part A 2015, spectroscopy. ACS Sens 2020, 5:423-430.
21:1982-1990.
82. Simon-Chica A, Fernández MC, Wülfers EM, Lother A, Hilgendorf I,
70. Rouabhia M, Park H, Meng S, Derbali H, Zhang Z: Electrical Seemann G, Ravens U, Kohl P, Schneider-Warme F: Novel
stimulation promotes wound healing by enhancing dermal insights into the electrophysiology of murine cardiac
fibroblast activity and promoting myofibroblast macrophages: relevance of voltage-gated potassium channels.
transdifferentiation. PLOS One 2013, 8:e71660. Cardiovasc Res (3) 2021, 118:798-813, https://doi.org/10.1093/cvr/
cvab126
71. Ko UH, Choi J, Choung J, Moon S, Shin JH: Physicochemically
tuned myofibroblasts for wound healing strategy. Sci Rep 2019, 83. McCann FV, Cole JJ, Guyre PM, Russell JAG: Action potentials in
9:1-12. macrophages derived from human monocytes. Science 1983,
219:991-993.
72. Wang Y, Rouabhia M, Zhang Z: Pulsed electrical stimulation
benefits wound healing by activating skin fibroblasts through 84. Woehlck HJ, Mccann FV: Action potentials in human
the TGFβ1/ERK/NF-κB axis. Biochim Biophys Acta — Gen Subj macrophages are calcium spikes. Cell Biol Int Rep 1986,
1860, 2016:1551-1559. 10:517-525.

www.sciencedirect.com Current Opinion in Biotechnology 75(2022) 102710

You might also like