Elsevier

Journal of Controlled Release

Volume 194, 28 November 2014, Pages 138-147
Journal of Controlled Release

PLGA nanoparticles loaded with host defense peptide LL37 promote wound healing

https://doi.org/10.1016/j.jconrel.2014.08.016Get rights and content

Abstract

Wound treatment remains one of the most prevalent and economically burdensome healthcare issues in the world. Poly (lactic-co-glycolic acid) (PLGA) supplies lactate that accelerates neovascularization and promotes wound healing. LL37 is an endogenous human host defense peptide that modulates wound healing and angiogenesis and fights infection. Hence, we hypothesized that the administration of LL37 encapsulated in PLGA nanoparticles (PLGA-LL37 NP) promotes wound closure due to the sustained release of both LL37 and lactate. In full thickness excisional wounds, the treatment with PLGA-LL37 NP significantly accelerated wound healing compared to PLGA or LL37 administration alone. PLGA-LL37 NP-treated wounds displayed advanced granulation tissue formation by significant higher collagen deposition, re-epithelialized and neovascularized composition. PLGA-LL37 NP improved angiogenesis, significantly up-regulated IL-6 and VEGFa expression, and modulated the inflammatory wound response. In vitro, PLGA-LL37 NP induced enhanced cell migration but had no effect on the metabolism and proliferation of keratinocytes. It displayed antimicrobial activity on Escherichia coli. In conclusion, we developed a biodegradable drug delivery system that accelerated healing processes due to the combined effects of lactate and LL37 released from the nanoparticles.

Introduction

Wound treatment and its medical complications remain one of the most prevalent and economically challenging healthcare issues in the world. The annual wound care products market has reached to $15.3 billion by 2010 and still the need for post-surgical wound care is sharply on the rise. In the USA alone there are more than 100 million wounds annually, including surgical incisions, trauma, burns, blast injuries, diabetic ulcers, ostomies, bedsores, and more [1], [2]. Acute wounds heal in a very orderly, timely and efficient manner characterized by four distinct, strictly connected but overlapping phases: hemostasis, inflammation, proliferation and remodeling. Any disturbances in these phases may result in an incomplete and improper restoration of the injured tissue [3]. In these severe clinical conditions wound repair may fail to proceed, leading to chronic non-healing wounds. Current treatment options are limited, costly, and inefficient. As a result, the development of new therapeutics is absolutely necessary to satisfy the unmet clinical need.

One of the simple and pragmatic solutions to faster wound healing process was the application of exogenous lactate that accelerates angiogenesis, activation of procollagen factors and recruitment of endothelial progenitor cells in wounds. Utilization of poly (lactic-co-glycolic acid) (PLGA) is one of the strategies to supply lactate sustainably [4]. Moreover, PLGA is biodegradable, biocompatible, has versatile degradation kinetics and approved by the European Medical Agency and Food and Drug Administration as an excipient for parenteral products [5]. Thus the key advantage of PLGA drug delivery systems is the polymer can perform the dual roles: being a wound healing agent itself and its capability to release loaded drugs sustainably to the wound.

LL37 belongs to antimicrobial peptides/host defense peptides that are part of the innate immune system and represent the first line of defense against many invading pathogens [6]. hCAP-18/LL37 is up to now the only antimicrobial peptide of the cathelicidin family identified in humans. LL37 has been detected in an inactive proform in several types of cells such as neutrophils, mast cells, monocytes, macrophages, NK cells, γδ cells, B cells, dermal epithelial cells and keratinocytes, inflamed skin and blister fluids. Any infection or tissue and cell damage that occurs during the wound may activate TLR(s) and/or an alteration in the cytokine milieu, which provides a trigger that activates the cell to degranulate. This leads to the release of the inactive hCAP18 precursor in the extracellular environment, where it can be processed by specific proteases into the active 37 amino acid long LL37 peptide [7], [8], [9]. LL37 exerts different immunomodulatory functions like broad antimicrobial activity, antiviral and antifungal activity, endotoxin-binding properties, modulation of pro-inflammatory response, chemotaxis, influence on cell proliferation and differentiation, promotion of wound healing and angiogenesis, etc. [10]. The major difficulty associated with LL37 administration is its immediate degradation in the wound environment and thus the treatment requires high dose and dosing frequency of LL37 [11] or gene therapy [12] to produce the therapeutic effect.

PLGA nanoparticles (NP) have been successfully proved to be efficient carriers for large biomolecules such as vaccines and proteins for the treatment of various ailments [5], [13]. Hence, we hypothesized that the administration of LL37 encapsulated in PLGA nanoparticles (PLGA-LL37 NP) could efficiently deliver the LL37 and accelerate wound closure through different mechanisms due to the encapsulated LL37 and lactate released from PLGA. Thus, we present the PLGA-LL37 NP for the active healing of dermal wounds and study their mechanisms of action.

Section snippets

PLGA-LL37 nanoparticle preparation

LL37 loaded PLGA (50:50, Mw 7000–17,000, acid terminated, Boehringer Ingelheim GmbH, DE) nanoparticles were prepared by W/O/W emulsion–solvent evaporation technique with a few modifications from the literature [14]. Briefly, 20 mg of PLGA polymer was dissolved in 1 ml dichloromethane (HPLC grade, Sigma-Aldrich, DE). 20 μg of LL37 (95.0% pure, Caslo ApS, DK) (peptide sequence LLGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES, theoretical molecular mass 4493.37) in 20 μl sterile water, was added to PLGA solution

Preparation, characterization, release and stability studies of PLGA-LL37 NP

PLGA-LL37 NP were prepared by double (W/O/W) emulsion–solvent evaporation technique using PVA as stabilizer. The size, PDI, zeta potential and encapsulation efficiency of PLGA-LL37-NP are presented in Table 1.

PLGA-LL37 NP were stable and showed no significant differences in properties such as size, zeta potential and drug loading (quantified by LC-ESI-MS) after 30 days of storage at room temperature (Table 1 of supplementary data 2). The in vitro drug release profile of LL37 from PLGA-LL37 NP

Discussion

Various delivery systems have been developed to provide controlled release of proteins and peptides. However, clinical translation of these systems has been discouraged by their drawbacks such as poor loading and low biocompatibility of delivery systems. But, PLGA NP protect the loaded cargo from external environment and thereby enhance its availability and activity. Particularly its active involvement in wound healing processes is highly appropriate in the choice of delivery systems for wound

Conclusions

Administration of LL37 in PLGA nanoparticles and combined therapeutic activities of PLGA and LL37 to fasten dermal wound healing were investigated in the current study (Scheme 1). We demonstrated that the PLGA-based sustained delivery of LL37 significantly improved the wound healing activity compared to PLGA or LL37 alone. The healing effect of PLGA-LL37 NP included higher re-epithelialization, granulation tissue formation and immunomodulation. PLGA-LL37 NP significantly up-regulated IL-6 and

Acknowledgments

The financial support (grant agreement number 289454) from the European Commission and Marie Curie Actions is greatly appreciated. We express our earnest thanks to Mr. Bernard Ucakar (Louvain Drug Research Institute, UCL, BE) and Mr. Raoul Rozenberg (Institute of Condensed Matter and Nanosciences, UCL, BE) for their help with animal experiments and mass spectrometry measurements respectively. Kiran Kumar Chereddy, Michela Comune and Claudia Moia are early stage researchers (ESR) of FP7 Marie

References (37)

  • M.D. Blanco et al.

    Development and characterization of protein-loaded poly(lactide-co-glycolide) nanospheres

    Eur. J. Pharm. Biopharm.

    (1997)
  • S. Fredenberg et al.

    The mechanisms of drug release in poly(lactic-co-glycolic acid)-based drug delivery systems—a review

    Int. J. Pharm.

    (2011)
  • R.M. Gallucci et al.

    Interleukin 6 indirectly induces keratinocyte migration

    J. Investig. Dermatol.

    (2004)
  • T.K. Hunt et al.

    Anaerobic metabolism and wound healing: an hypothesis for the initiation and cessation of collagen synthesis in wounds

    Am. J. Surg.

    (1978)
  • O. Sørensen et al.

    The human antibacterial cathelicidin, hCAP-18, is synthesized in myelocytes and metamyelocytes and localized to specific granules in neutrophils

    Blood

    (1997)
  • M. Carretero et al.

    In vitro and in vivo wound healing-promoting activities of human cathelicidin LL-37

    J. Investig. Dermatol.

    (2008)
  • M.G. Franz et al.

    Guidelines to aid healing of acute wounds by decreasing impediments of healing

    Wound Repair Regen.

    (2008)
  • C.K. Sen et al.

    Human skin wounds: a major and snowballing threat to public health and the economy

    Wound Repair Regen.

    (2009)
  • Cited by (0)

    1

    Equal contribution.

    View full text