Regenerative Nanomedicine (Ed. A. Seifalian)
Magnetic field-guided cell delivery with nanoparticle-loaded human corneal endothelial cells

https://doi.org/10.1016/j.nano.2014.12.002Get rights and content

Abstract

To improve the delivery and integration of cell therapy using magnetic cell guidance for replacement of corneal endothelium, here we assess magnetic nanoparticles’ (MNPs’) effects on human corneal endothelial cells (HCECs) in vitro. Biocompatible, 50 nm superparamagnetic nanoparticles endocytosed by cultured HCECs induced no short- or long-term change in viability or identity. Assessment of guidance of the magnetic HCECs in the presence of different magnet shapes and field strengths showed a 2.4-fold increase in delivered cell density compared to gravity alone. After cell delivery, HCECs formed a functional monolayer, with no difference in tight junction formation between MNP-loaded and control HCECs. These data suggest that nanoparticle-mediated magnetic cell delivery may increase the efficiency of cell delivery without compromising HCEC survival, identity or function. Future studies may assess the safety and efficacy of this therapeutic modality in vivo.

From the Clinical Editor

The authors show in this article that magnetic force facilitates the delivery of human corneal endothelial cells loaded by superparamagnetic nanoparticles to cornea, without changing their morphology, identity or functional properties. This novel idea can potentially have vast impact in the treatment of corneal endothelial dystrophies by providing self-endothelial cells after ex-vivo expansion.

Graphical abstract

One of the main challenges in cell therapy is the targeting of the cells to the right place. We assessed the effect of adding magnetic nanoparticles (MNPs) to human corneal endothelial cells (HCECs) in culture. In the presence of a magnetic field, MNP-loaded HCEC delivered cell density increased 2.4-fold compared to gravity alone. We also found no short- or long-term changes in cell viability and identity, or in HCECs’ ability to form a tight, functional monolayer. Thus, nanoparticle-mediated cell delivery may be a viable solution for corneal cell therapy.

  1. Download : Download high-res image (54KB)
  2. Download : Download full-size image

Section snippets

Cell culture

Cadaveric donor corneas, preserved at 4 °C in Optisol-GS (Baush & Lomb, Rochester, NY), were obtained from the Lions Eye Institute for Transplant and Research (Tampa, FL), the Florida Lions Eye Bank (Miami, FL) and the National Disease Research Interchange (NDRI, Philadelphia, PA). Primary cultures of HCECs were purified and expanded following the method described by Zhu and Joyce (2004) with some modifications. In brief, corneas were rinsed 3 times in M199 with gentamicin 50 μg/μl

Viability and movement of MNP-loaded HCECs in vitro

Since magnetic nanoparticles could be potentially used in cell therapy to help direct HCECs to the corneal endothelium, we first asked whether adding increasing volumes of 50 nm diameter superparamagnetic nanoparticles (MNPs) is toxic to HCECs in culture. There was no difference in viability, by Trypan Blue exclusion, between control HCECs and HCECs incubated with 10, 20, 100, or 1000 μL of MNPs after 1 day in vitro (DIV) (Figure 1, A-B).To determine whether treating HCECs with MNPs renders the

Discussion

Here we find that nanoparticle-endocytosing HCECs show no difference in viability, identity, or barrier function compared to untreated cells. Nanotechnology use in medicine has expanded tremendously over the last two decades. Nanomaterials are currently used clinically to augment the potency of prostate cancer therapy, to enhance imaging modalities, and to improve drug bioavailability, among other applications.24, 25, 26, 27, 28, 29, 30 Nanoparticle drug delivery systems have been designed to

Acknowledgment

We are indebted to Ashley Morganti at the Lions Eye Institute for Transplant and Research (Tampa, FL) and to the San Diego Eye Bank for providing human corneas of remarkable quality for research. We thank colleagues at the University of Miami Peggy Bates and Gabriel Gaidosh for their assistance with transmission and confocal microscopy, Bill Feuer for his expertise in statistics, and Alex Kreymerman and Daniel Wolfgang Pita-Thomas (now at Washington University, St Louis, MO) for scientific

References (51)

  • S.P. Chawla et al.

    Phase I/II and phase II studies of targeted gene delivery in vivo: intravenous Rexin-G for chemotherapy-resistant sarcoma and osteosarcoma

    Mol Ther

    (2009)
  • W.M. Bourne

    Biology of the corneal endothelium in health and disease

    Eye (Lond)

    (2003)
  • F.W. Price et al.

    Evolution of endothelial keratoplasty

    Cornea

    (2013)
  • Y. Shimazono

    The state of the international organ trade: a provisional picture based on integration of available information

    Bull World Health Organ

    (2007)
  • World_Health_Organization

    Second Global Consultation on Regulatory Requirements for Human Cells and Tissues for Transplantation — towards global harmonization through graduated standards

    (2006)
  • M.O. Price et al.

    Endothelial keratoplasty — a review

    Clin Experiment Ophthalmol

    (2010)
  • K.H. Chen et al.

    Transplantation of adult human corneal endothelium ex vivo: a morphologic study

    Cornea

    (2001)
  • G.S. Peh et al.

    Human corneal endothelial cell expansion for corneal endothelium transplantation: an overview

    Transplantation

    (2011)
  • N.C. Joyce et al.

    Human corneal endothelial cell proliferation: potential for use in regenerative medicine

    Cornea

    (2004)
  • G.S. Peh et al.

    Cultivation of human corneal endothelial cells isolated from paired donor corneas

    PLoS One

    (2011)
  • K. Alvarez-Delfin et al.

    A cell therapy approach to address corneal endothelial dysfunction

  • B.M. Stramer et al.

    Monoclonal antibody (3G5)-defined ganglioside: cell surface marker of corneal keratocytes

    Invest Ophthalmol Vis Sci

    (2004)
  • A.S. Jun et al.

    Microarray analysis of gene expression in human donor corneas

    Arch Ophthalmol

    (2001)
  • M. Bohnke et al.

    Detection of neurone-specific enolase in long-term cultures of human corneal endothelium

    Graefes Arch Clin Exp Ophthalmol

    (1998)
  • M.M. Stiemke et al.

    Maturation of the corneal endothelial tight junction

    Invest Ophthalmol Vis Sci

    (1991)
  • Cited by (35)

    • Corneal endothelial transplantation from bench to bedside: A review of animal models and their translational value for therapeutic development

      2022, Experimental Eye Research
      Citation Excerpt :

      By contrast, an “off-the-shelf” and injectable cellular therapeutic could revolutionize the endothelial cell replacement space and offer ophthalmologists, cornea and non-cornea trained alike, a treatment tool with dramatically expanded options for administration. Multiple groups have cultured CECs for delivery on various scaffolds, including collagen sheets, amniotic membranes, and even donor Descemet membrane (Ishino et al., 2004; Joyce and Zhu, 2004; Mimura et al., 2004a; Moysidis et al., 2015; Niu et al., 2014; Ozcelik et al., 2013; Peh et al., 2017; Watanabe et al., 2011). Attempts to deliver cultured cells intracamerally without a scaffold have also been explored; Shigeru Kinoshita's team has been most successful in facilitating a microenvironment that suitably accomplishes this while achieving long-term corneal transparency (Kayukawa et al., 2020; Kinoshita et al., 2018; Mimura et al., 2003; Okumura et al., 2012; Schlötzer-Schrehardt et al., 2021).

    • Approaches for corneal endothelium regenerative medicine

      2022, Progress in Retinal and Eye Research
      Citation Excerpt :

      Co-delivery of the cells with the ROCK inhibitor Y-27632 combined with prone position of the recipient significantly improved cell adhesion (Okumura et al, 2012, 2014b; Peh et al., 2019). Another strategy explored the enhancement of CEC attachment by the use of magnetic force using cells laden with ferromagnetic beads (Mimura et al., 2003; Moysidis et al., 2015; Patel et al., 2009). In the pioneering first-in-human clinical trial using primary cultured CECs, Kinoshita and colleagues reversed corneal edema in cataract surgery–derived bullous keratopathy patients and FECD patients with an injection of primary cultured CECs together with Y-27632 (Kinoshita et al., 2018).

    • Microfluidic mechanoporation for cellular delivery and analysis

      2022, Materials Today Bio
      Citation Excerpt :

      The methods are advantageous because of minimal chances of inducing harsh cellular responses such as unwanted immunogenic or chemical reactions, thereby improving delivery efficiencies. Electroporation [13–21], optoporation [22–28], magnetoporation [29–31], acoustoporation [32–35] and mechanoporation [36–60] are some of the most popular membrane disruption techniques. Although each of these methods have their own distinct features (elucidated in detail in Table 2), all of these, except mechanoporation, require an external energy field for membrane permeabilization.

    • Corneal endothelial dysfunction: Evolving understanding and treatment options

      2021, Progress in Retinal and Eye Research
      Citation Excerpt :

      Delivery of the cultured cells in animal models has followed three main strategies: cell injection therapy, ultra-thin monolayer cell sheets, or cell-based carrier systems (Parikumar et al., 2018). Cell injection therapy involves the direct delivery of endothelial cells by intracameral injection in combination with ferromagnetic induction (Moysidis et al., 2015), prone posture (Okumura et al., 2012), augmentation with Rock inhibitors, or a combination of all of these methods (Xia et al., 2019) (Fig. 16). The appeal of cell injection therapy is the simplistic nature of the procedure, however results in animal studies have varied between different groups (Bostan et al., 2016; Okumura et al., 2016a).

    • Corneal replacement tissue

      2020, Principles of Tissue Engineering
    View all citing articles on Scopus

    Disclosure of Commercial Relationship(s): Emmetrope Ophthalmics LLC: SNM (Consultant, Patent), NJK (Employee, Patent), JLG (Board Member, Patent). All other authors have no relevant commercial relationships or financial interests to disclose.

    We gratefully acknowledge funding from the NEI (P30-EY022589, UCSD and P30-EY014801, University of Miami), and an unrestricted grant from Research to Prevent Blindness, Inc.

    View full text