Elsevier

Journal of Controlled Release

Volume 339, 10 November 2021, Pages 473-483
Journal of Controlled Release

Amphiphilic galactomannan nanoparticles trigger the alternative activation of murine macrophages

https://doi.org/10.1016/j.jconrel.2021.10.017Get rights and content

Abstract

Macrophages are highly plastic phagocytic cells that can exist in distinct phenotypes and play key roles in physiological and pathological pathways. They can be classically activated to the pro-inflammatory M1 phenotype or alternatively activated to an M2 anti-inflammatory one by various stimuli in the biological milieu. Different biomaterials polarize macrophages to M1 or M2 phenotypes and emerged as a very promising strategy to modulate their activation and performance. In this work, we investigate the ability of drug-free amphiphilic nanoparticles (hydrodynamic diameter of ~130 nm) produced by the self-assembly of a graft copolymer of hydrolyzed galactomannan, a natural polysaccharide of galactose and mannose, that was hydrophobized in the side-chain with poly(methyl methacrylate) blocks and that can encapsulate hydrophobic drugs, to trigger macrophage polarization. The compatibility and uptake of the nanoparticles are demonstrated in the murine macrophage cell line RAW264.7 by a metabolic assay, confocal laser scanning fluorescence microscopy (CLSFM) and imaging flow cytometry in the absence and the presence of endocytosis inhibitors. Results indicate that they are internalized by both clathrin- and caveolin-mediated endocytosis. The ability of these drug-free nanoparticles to polarize these cells to the M2-like phenotype and to switch an M1 to an M2 phenotype is confirmed by the downregulation of the M1 marker cluster of differentiation 80 (CD80), and upregulation of M2 markers CD163 and CD206, as measured by flow cytometry and CLSFM. In addition, we preliminarily assess the effect of the nanoparticles on wound healing by tracking the closure of an artificial wound of RAW264.7 macrophages in a scratch assay. Findings indicate a faster closure of the wound in the presence of the nanoparticles with respect to untreated cells. Finally, a migration assay utilizing a macrophage/fibroblast co-culture model in vitro demonstrates that M2 polarization increases fibroblast migration by 24-fold with respect to untreated cells. These findings demonstrate the ability of this nanotechnology platform to interfere and change the macrophages phenotype in vitro and represent robust evidence for the investigation of their therapeutic performance alone or in combination with an encapsulated hydrophobic drug in wound models in vivo

Introduction

Macrophages are highly plastic cells of the immune system that play a key roles in the innate and the adaptive immune response and the normal tissular and organic development and function [1]. They are a source of growth factors [2], recruit other immune cells, and are involved in different pathophysiological conditions [[3], [4], [5], [6], [7], [8]]. Macrophages display a wide range of phenotypes and change their physiological performance according to stimuli in the biological milieu [9,10]. Macrophage polarization is an in vitro model that defines two extreme phenotypes of the activation spectrum [11]. In this model, naïve macrophages (M0) can differentiate to a classically activated (M1) phenotype that interacts with T helper type 1 cells and releases pro-inflammatory factors such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Conversely, the alternatively activated (M2) phenotype mediates T helper type 2 cell response and releases anti-inflammatory cytokines (e.g., IL-4 and IL-10) that inhibit inflammation and promote tissue regeneration [12,13]. In vivo, macrophages can display intermediate polarization states and both phenotypes can co-exist [14]. In addition, macrophages can switch their phenotype from M1 to M2 and vice versa or maintain their M0 state in the absence of external stimuli. The M1 phenotype is involved in the response to bacterial and viral infections, eliminates aging or necrotic cells [15], and improves the direct killing of cancer cells [16]. Conversely, the M2 phenotype is commonly implicated in the response to fungi, parasites, and apoptotic cells [17] and is characterized by high expression of scavenging molecules, mannose and galactose receptors, high phagocytosis capacity, production of extracellular matrix, and stimulation of tissue remodeling, repair and regeneration [18].

The ability to trigger and direct macrophage polarization by small-molecule and biological drugs has emerged as a new therapeutic approach [19,20]. However, when these therapies are not targeted to macrophages, they might be accompanied by systemic side-effects on other cell types [21].

Biomaterials with different chemical, structural and mechanical properties and in different forms (e.g., particles, films) can be used as immunomodulators because they trigger macrophage polarization, provide a more controlled temporal and spatial release of key cytokines and growth factors and mimic better complex physiological signaling patterns with minimal toxicity [[22], [23], [24], [25]]. Due to their smaller size and larger surface-to-volume ratio, nanoparticles (NPs) have been extensively investigated in drug delivery and targeting and recently, proposed to tune macrophage polarization [14]. Amphiphilic polymeric NPs (e.g., polymeric micelles) are self-assembly nanostructures formed by the spontaneous aggregation of copolymeric amphiphiles in water above the critical aggregation concentration [26,27]. They display sizes between a few tens to several hundreds of nanometers and undergo differential biodistribution in body tissues and organs [26,27]. Their surface can be tailored to selectively bind receptors overexpressed by macrophages and undergo uptake by different pathways [28,29]. Lectin-like receptors are transmembrane proteins that selectively bind glucose and mannose and they are overexpressed by M2 macrophages [20,30]. Another key β-glucan receptor in macrophages is dectin-1 [31]. For example, particulate β-glucan switches the M2 phenotype to an M1-like one [32].

Galactomannans (GMs) are polysaccharides made of a mannose backbone and side galactose residues [33]. Polymeric NPs surface-modified with hydrolyzed galactomannan (hGM) improve the intracellular delivery of encapsulated drugs to macrophages [34]. Recently, we demonstrated that amphiphilic hGM-g-poly(methyl methacrylate) (PMMA) NPs efficiently encapsulate hydrophobic drugs (e.g., imatinib) and that their accumulation in pediatric patient-derived xenograft sarcoma models in mouse correlates with the expression of glucose transporter 1 [35].

In this conceptual framework, we hypothesized that the interaction and uptake of hGM-g-PMMA NPs by macrophages triggers their polarization. Thus, in this work, we first investigated the compatibility and endocytic mechanisms by which hGM-g-PMMA NPs (PMMA content of 30% w/w) are internalized by the murine macrophage cell line RAW264.7 by using a metabolic assay, confocal laser scanning fluorescence microscopy (CLSFM) and imaging flow cytometry. Then, the ability of these NPs to induce M2-polarization in vitro was demonstrated by flow cytometry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and enzyme-linked immunosorbent assay (ELISA). Next, their wound healing performance was assessed in an artificial wound model by a real time scratch assay. Finally, the ability of murine macrophages pre-exposed to drug-free hGM-g-PMMA NPs and polarized to a M2 phenotype to induce the migration of murine fibroblasts was studied in a macrophage/fibroblast co-culture model.

Section snippets

Materials

GM (from locust bean gum) was supplied by Glentham Life Sciences (Corsham, UK) and hydrolyzed under acid conditions to produce hGM and dialyzed before use [34,35]. Methyl methacrylate (MMA, Alfa Aesar, Heysham, UK) was distilled under vacuum at 35–40 °C to remove the free radical inhibitor. Cerium(IV) ammonium nitrate (CAN, Sigma-Aldrich, St. Louis, MO, USA), nitric acid 70% (Bio-Lab, Jerusalem, Israel) and tetramethylethylenediamine (TEMED, Alfa Aesar) were used without further purification.

Synthesis and characterization of the nanoparticles

An amphiphilic hGM-g-PMMA copolymer containing 30% w/w PMMA (hGM-PMMA30) was synthesized by the free radical graft polymerization of MMA in water [35]. The copolymer was solubilized in different physiologically relevant media (0.1% w/v) at a concentration above the critical aggregation concentration [35] to produce the NPs by self-assembly and the Dh and the PDI measured by DLS at 25 and 37 °C. In PBS, NPs display a monomodal size population (Dh of 128–130 nm, by intensity) and PDI of 0.14–0.50

Conclusions

Active drug targeting of macrophages by means of glycosylated nanoparticles that bind lectin-like receptors and other transmembrane receptors overexpressed by these immune cells has been extensively investigated. Conversely, the exploitation of the pathways to immunomodulate and polarize them has been remarkably scarce, especially with drug-free nanocarriers.

In this work, we investigated the polarization of murine macrophages by drug-free hGM-g-PMMA NPs. Imaging flow-cytometry and CLSFM

Declaration of Competing Interest

None.

Acknowledgments

A.S. thanks the financial support of Technion and the Russell Berrie Nanotechnology Institute (RBNI, Technion). We thank Nitzan Dahan and Efrat Barak at the Life Sciences and Engineering Infrastructure Center (Lorry I. Lokey Center, Technion) for technical assistance in CLSFM and imaging flow cytometry, respectively.

References (69)

  • X. Li et al.

    The systematic evaluation of size-dependent toxicity and multi-time biodistribution of gold nanoparticles

    Colloids Surf. B: Biointerfaces

    (2018)
  • J.A. Swanson et al.

    Macropinocytosis

    Trends Cell Biol.

    (1995)
  • H. Yadav et al.

    Research progress in galactomannan-based nanomaterials: synthesis and application

    Int. J. Biol. Macromol.

    (2020)
  • M. Kandušer et al.

    The temperature effect during pulse application on cell membrane fluidity and permeabilization

    Bioelectrochemistry

    (2008)
  • T. Kawamoto et al.

    TAK-242 selectively suppresses toll-like receptor 4-signaling mediated by the intracellular domain

    Eur. J. Pharmacol.

    (2008)
  • M.J. Feito et al.

    Characterization of M1 and M2 polarization phenotypes in peritoneal macrophages after treatment with graphene oxide nanosheets

    Colloids Surf. B: Biointerfaces

    (2019)
  • C.Y. Liu et al.

    M2-polarized tumor-associated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway

    Lab. Investig.

    (2013)
  • C.M. Minutti et al.

    Tissue-specific contribution of macrophages to wound healing

    Semin. Cell Dev. Biol.

    (2017)
  • J.W. Pollard

    Trophic macrophages in development and disease

    Nat. Rev. Immunol.

    (2009)
  • A. Chawla et al.

    Macrophage-mediated inflammation in metabolic disease

    Nat. Rev. Immunol.

    (2011)
  • J.M. Olefsky et al.

    Macrophages, inflammation, and insulin resistance

    Annu. Rev. Physiol.

    (2010)
  • W.J. Boyle et al.

    Osteoclast differentiation and activation

    Nature

    (2003)
  • T.A. Wynn et al.

    Macrophages: master regulators of inflammation and fibrosis

    Semin. Liver Dis.

    (2010)
  • K.S. Jones

    Biomaterials as vaccine adjuvants

    Biotechnol. Prog.

    (2008)
  • N.A. Hotaling et al.

    Biomaterial strategies for immunomodulation

    Annu. Rev. Biomed. Eng.

    (2015)
  • P.J. Murray

    Macrophage polarization

    Annu. Rev. Physiol.

    (2017)
  • S.K. Biswas et al.

    Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm

    Nat. Immunol.

    (2010)
  • G. Lopez-Castejón et al.

    Novel macrophage polarization model: from gene expression to identification of new anti-inflammatory molecules

    Cell. Mol. Life Sci.

    (2011)
  • G. Genard et al.

    Proton irradiation orchestrates macrophage reprogramming through NFκB signaling

    Cell Death Dis.

    (2018)
  • N.N. Parayath et al.

    Repolarization of tumor-associated macrophages in a genetically engineered nonsmall cell lung cancer model by intraperitoneal administration of hyaluronic acid-based nanoparticles encapsulating MicroRNA-125b

    Nano Lett.

    (2018)
  • A. Sica et al.

    Macrophage plasticity and polarization: in vivo veritas

    J. Clin. Invest.

    (2012)
  • R. Garash et al.

    Drug delivery strategies to control macrophages for tissue repair and regeneration

    Exp. Biol. Med.

    (2016)
  • D. Reichel et al.

    Biological effects of nanoparticles on macrophage polarization in the tumor microenvironment

    Nanotheranostics

    (2019)
  • X. Xie et al.

    The effect of shape on cellular uptake of gold nanoparticles in the forms of stars, rods, and triangles

    Sci. Rep.

    (2017)
  • Cited by (15)

    • The immunomodulatory activity of degradation products of Sesbania cannabina galactomannan with different molecular weights

      2022, International Journal of Biological Macromolecules
      Citation Excerpt :

      For example, the expression of mannose receptor and mannose-binding lectins is known to correlate with mannose-containing polysaccharide recognition by macrophages and the activation of downstream signaling cascades [53]. This may require the use of other receptor inhibitors for research, such as chlorpromazine, flipin or cytochalasin B [54]. Based on these results, we found that the molecular weight may be not directly related the immunomodulatory activity of GM, while the hydroxyl group does.

    View all citing articles on Scopus
    View full text