Elsevier

Brain Research Bulletin

Volume 152, October 2019, Pages 265-284
Brain Research Bulletin

Review
Neural responses to electrical stimulation in 2D and 3D in vitro environments

https://doi.org/10.1016/j.brainresbull.2019.07.016Get rights and content

Highlights

  • Robust review of the methods of electrical stimulation.

  • Discussion of the influence of electrical stimulation on neural cells.

  • Influence of biomaterials and 2D vs 3D environments on neural response to ES.

  • Mechanisms of neural cell response to ES remain a challenging opportunity.

Abstract

Electrical stimulation (ES) to manipulate the central (CNS) and peripheral nervous system (PNS) has been explored for decades, recently gaining momentum as bioelectronic medicine advances. The application of ES in vitro to modulate a variety of cellular functions, including regenerative potential, migration, and stem cell fate, are being explored to aid neural degeneration, dysfunction, and injury. This review describes the materials and approaches for the application of ES to the PNS and CNS microenvironments, towards an improved understanding of how ES can be harnessed for beneficial clinical applications. Emphasized are some recent advances in ES, including conductive polymers, methods of charge transfer, impact on neural cells, and a brief overview of alternative methodologies for cellular targeting including magneto, ultrasonic, and optogenetic stimulation. This review will examine how heterogenous cell populations, including neurons, glia, and neural stem cells respond to a wide range of conductive 2D and 3D substrates, stimulation regimes, known mechanisms of response, and how cellular sources impact the response to ES.

Introduction

According to the U.S Department of Health and Human Services over 20 million people suffer or have suffered from peripheral nerve damage (Services 2014). This damage compromises the physical integrity of the Peripheral Nervous System (PNS), impacting the bi-directional transmission of electrical signals from limbs and organs to the Central Nervous System (CNS). Patients with peripheral nerve injuries (PNIs) present symptoms that involve numbness, loss of sensation, and even mechanical dysfunction resulting in lost productivity and increased socioeconomic isolation and burden. Minor and non-localized injuries with low risk of functional loss (e.g. nerve damage caused by diabetes) are generally not treated, relying on the patient’s self-regeneration capacities for recovery. On the other hand, severe nerve damage, e.g. traumatic injuries that compromise the nerve continuity, often require clinical interventions that represent around 200,000 repairs annually (Kehoe et al., 2012). Unfortunately, in these cases only 50% of patients regain useful function and few will completely recover resulting in long term disabilities, thus motivating the need for new and rationally defined treatment options.

Further, in the CNS approximately 1.7 million U.S. citizens suffer from traumatic brain injuries (TBIs) with an economic burden of up to $76.5 billion (Faul et al., 2010; Coronado et al., 2010, 2012; Cancelliere et al., 2017). In addition to TBI, there are 12,000–20,000 new spinal cord injuries (SCIs) per year (Raspa et al., 2016; Mazzoleni et al., 2013; Sahni and Kessler, 2010; Jazayeri et al., 2015). With the inclusion of neurodegenerative disorders, such as Parkinson’s and Alzheimer’s disease that also have limited therapeutic interventions, the impact of neural dysfunction on the quality of life for millions of people remains limited. In the CNS, regenerative capacity is limited by inhibitory barriers, such as glial scar formation and immune cell-mediated inflammation, motivating new therapeutic approaches such as electrical stimulation (ES) to modulate cellular function for improved restoration of function (Grinsell and Keating, 2014).

The application of ES for biological applications has been examined for over two hundred years, with increasing interest as technology and methods for cell culture and interfacing have expanded (Bresadola, 1998; Galvani, 1791). It is known that endogenous electrical cues drive development and morphogenesis, cellular communication networks, and neuronal and glial cellular signaling (Baer and Colello, 2016; Funk, 2015; Adams and Levin, 2013; Cao et al., 2013; Nuccitelli, 2003; Hotary and Robinson, 1992; Stump and Robinson, 1983); thus harnessing native machinery to modulate cellular activity in a controlled manner is attractive. ES has shown promise in modulating neural cell morphology (Shen et al., 2016; Yan et al., 2014; Huang et al., 2012b; Koppes et al., 2011; Lee et al., 2009) and impacting neural stem cell (NSC) fate (George et al., 2017; Hassarati et al., 2016; Cao et al., 2013; Jahanshahi et al., 2013; Park et al., 2011). However, a main limitation in successful translation to the clinic is the varied parameters, materials, and heterogeneous cell populations that limit a defined and mechanistic understanding of how to best modulate neural and support cells for specific therapeutic advantages.

This review focuses on the impact of ES in vitro, in 2D and 3D microenvironments to modulate neural outgrowth and function, support cell (e.g. glia), and NSC behavior. A consideration for how ES is applied, which materials are utilized for cellular and culture media interfacing, as well as limitations in mechanistic understanding is discussed. A mechanistic understanding will enable more targeted approaches in ES for specific cellular responses within heterogenous tissues to be realized. A brief outlook on alternative stimulation regimes, such as mechanical and light stimulation is also introduced. For some further reading on in vivo stimulation, readers are directed elsewhere (Koppes et al., 2014a; Ren et al., 2016; Wenger et al., 2016; Young, 2015; Angeli et al., 2014; Beuter et al., 2014; Kalia et al., 2013; Miocinovic et al., 2013; Hardenacke et al., 2012; Deuschl et al., 2011).

Section snippets

Historical overview

The link between electricity and biology began with the work of Luigi Galvani in the 18th century (Bresadola, 1998). By eliciting muscle contractions via ES of the sciatic nerve in a frog post mortem, Galvani was the first to demonstrate the excitability of nervous tissue. The influence of an applied electric field (EF) has been well documented for polarized cell populations including neurons, muscle, and glia, but all cells may be influenced by charge and ionic gradients (Batista Napotnik et

Neurons

Neurons synapse with other neurons to form neural networks, muscles to control movement, or convert sensory input intro electrochemical signals. A fundamental property of neurons is their ability to plastically respond to endogenous and exogenous stimuli (Sasmita et al., 2018; Lu et al., 2019; Werner and Stevens, 2015). Recent advances identified a plethora of neuronal subpopulations characterized by regional and epigenetic differences granting fine-tuned control over function (Lake et al., 2016

Future directions and other modes of stimulation

In addition to direct electrical coupling, a number of strategies for indirect coupling are being implemented to apply stimulation to in vitro cultures. These types of mechanisms will enable targeting of specific areas or spatiotemporal control over specific subpopulations of cells, enabling a minimization of side effects, off site targeting, or electrode byproducts at the site of application. An electromagnetic field can be induced via two parallel plates in close proximity (0.5–2 mm) to

Conclusions

Over the past few decades, the application of exogenous ES to modulate neural cell behavior for regenerative and therapeutic applications has gained momentum. Bioelectrical communication between neurons and support cell populations for restoration of function and wound healing has great potential for aiding recovery and regeneration, however there are still hurdles that require attention for the clinical relevance to be realized. As described above, the cell type and heterogeneity, species and

Declaration of Competing Interest

Conflict of Interest and Authorship Conformation Form Please check the following as appropriate:

X All authors have participated in (a) conception and design, or analysis and interpretation of the data; (b) drafting the article or revising it critically for important intellectual content; and (c) approval of the final version.

X This manuscript has not been submitted to, nor is under review at, another journal or other publishing venue.

X The authors have no affiliation with any organization with

References (232)

  • L. Dulabon et al.

    Reelin binds alpha3beta1 integrin and inhibits neuronal migration

    Neuron

    (2000)
  • Leandro Forciniti et al.

    Schwann cell response on polypyrrole substrates upon electrical stimulation

    Acta Biomater.

    (2014)
  • P.M. George et al.

    Electrical preconditioning of stem cells with a conductive polymer scaffold enhances stroke recovery

    Biomaterials

    (2017)
  • I. Goswami et al.

    Influence of pulsed electric fields and mitochondria-cytoskeleton interactions on cell respiration

    Biophys. J.

    (2018)
  • S. Grehl et al.

    Cellular and molecular changes to cortical neurons following low intensity repetitive magnetic stimulation at different frequencies

    Brain Stimul.

    (2015)
  • A. Gritli-Linde et al.

    The whereabouts of a morphogen: direct evidence for short- and graded long-range activity of hedgehog signaling peptides

    Dev. Biol. (Basel)

    (2001)
  • G.J. Harry

    Microglia during development and aging

    Pharmacol. Ther.

    (2013)
  • H. Hayashi et al.

    The effect of pulsed electric fields on the electrotactic migration of human neural progenitor cells through the involvement of intracellular calcium signaling

    Brain Res.

    (2016)
  • K.B. Hotary et al.

    Endogenous electrical currents and the resultant voltage gradients in the chick embryo

    Dev. Biol. (Basel)

    (1990)
  • M.R. Abidian et al.

    Conducting-polymer nanotubes improve electrical properties, mechanical adhesion, neural attachment, and neurite outgrowth of neural electrodes

    Small

    (2010)
  • M.R. Abidian et al.

    Interfacing conducting polymer nanotubes with the central nervous system: chronic neural recording using poly(3,4-ethylenedioxythiophene) nanotubes

    Adv. Mater.

    (2009)
  • D.S. Adams et al.

    Endogenous voltage gradients as mediators of cell-cell communication: strategies for investigating bioelectrical signals during pattern formation

    Cell Tissue Res.

    (2013)
  • S. Ahadian et al.

    Hybrid hydrogels containing vertically aligned carbon nanotubes with anisotropic electrical conductivity for muscle myofiber fabrication

    Sci. Rep.

    (2014)
  • O. Akhavan et al.

    Differentiation of human neural stem cells into neural networks on graphene nanogrids

    J. Mater. Chem. B

    (2013)
  • N.A. Alba et al.

    In vivo electrochemical analysis of a PEDOT/MWCNT neural electrode coating

    Biosensors (Basel)

    (2015)
  • John K. Alexander et al.

    Electric field-induced astrocyte alignment directs neurite outgrowth

    Neuron Glia Biol.

    (2006)
  • M.A. Anderson et al.

    Astrocyte scar formation aids central nervous system axon regeneration

    Nature

    (2016)
  • M.A. Anderson et al.

    Required growth facilitators propel axon regeneration across complete spinal cord injury

    Nature

    (2018)
  • C.A. Angeli et al.

    Altering spinal cord excitability enables voluntary movements after chronic complete paralysis in humans

    Brain

    (2014)
  • C.A. Ariza et al.

    The influence of electric fields on hippocampal neural progenitor cells

    Stem Cell Rev.

    (2010)
  • M. Arocena et al.

    A time-lapse and quantitative modelling analysis of neural stem cell motion in the absence of directional cues and in electric fields

    J. Neurosci. Res.

    (2010)
  • R. Babona-Pilipos et al.

    Biphasic monopolar electrical stimulation induces rapid and directed galvanotaxis in adult subependymal neural precursors

    Stem Cell Res. Ther.

    (2015)
  • M.L. Baer et al.

    Endogenous bioelectric fields: a putative regulator of wound repair and regeneration in the central nervous system

    Neural Regen. Res.

    (2016)
  • J. Banerjee et al.

    Immediate effects of repetitive magnetic stimulation on single cortical pyramidal neurons

    PLoS One

    (2017)
  • A. Barateiro et al.

    Oligodendrocyte development and myelination in neurodevelopment: molecular mechanisms in health and disease

    Curr. Pharm. Des.

    (2016)
  • B.A. Barres et al.

    Proliferation of oligodendrocyte precursor cells depends on electrical activity in axons

    Nature

    (1993)
  • M.J. Barton et al.

    The glia response after peripheral nerve injury: a comparison between schwann cells and olfactory ensheathing cells and their uses for neural regenerative therapies

    Int. J. Mol. Sci.

    (2017)
  • E. Basham et al.

    Circuit and coil design for in-vitro magnetic neural stimulation systems

    IEEE Trans. Biomed. Circuits Syst.

    (2009)
  • Bazargani

    Narges, and David %J nature neuroscience Attwell

    (2016)
  • S.J. Beebe et al.

    Diverse effects of nanosecond pulsed electric fields on cells and tissues

    DNA Cell Biol.

    (2003)
  • R.B. Borgens

    Voltage gradients and ionic currents in injured and regenerating axons

    Adv. Neurol.

    (1988)
  • R.B. Borgens et al.

    Functional recovery after spinal cord hemisection in guinea pigs: the effects of applied electric fields

    J. Comp. Neurol.

    (1990)
  • R.B. Borgens et al.

    Delayed application of direct current electric fields in experimental spinal cord injuries

    Restor. Neurol. Neurosci.

    (1993)
  • R.B. Borgens et al.

    Effects of applied electric fields on clinical cases of complete paraplegia in dogs

    Restor. Neurol. Neurosci.

    (1993)
  • E.S. Boyden et al.

    Millisecond-timescale, genetically targeted optical control of neural activity

    Nat. Neurosci.

    (2005)
  • S.B. Brummer et al.

    Electrochemical considerations for safe electrical stimulation of the nervous system with platinum electrodes

    IEEE Trans. Biomed. Eng.

    (1977)
  • A. Canales et al.

    MUltifunctional fibers for simultaneous optical, electrical and chemical interrogation of neural circuits in vivo

    Nat. Biotechnol.

    (2015)
  • Carol Cancelliere et al.

    Epidemiology of isolated versus nonisolated mild traumatic brain injury treated in emergency departments in the United States, 2006-2012: sociodemographic characteristics

    J. Head Trauma Rehabil.

    (2017)
  • C. Cannizzaro et al.

    Practical aspects of cardiac tissue engineering with electrical stimulation

    Methods Mol. Med.

    (2007)
  • L. Cao et al.

    Endogenous electric currents might guide rostral migration of neuroblasts

    EMBO Rep.

    (2013)
  • Cited by (0)

    View full text