Elsevier

Biomaterials

Volume 33, Issue 27, September 2012, Pages 6559-6569
Biomaterials

Simultaneous induction of autophagy and toll-like receptor signaling pathways by graphene oxide

https://doi.org/10.1016/j.biomaterials.2012.05.064Get rights and content

Abstract

Graphene oxide (GO) nanosheets have sparked growing interests in biological and medical applications. This study examined how macrophage, the primary immune cell type engaging microbes, responded to GO treatment. We uncovered that incubation of macrophage cell RAW264.7 with GO elicited autophagy in a concentration-dependent manner, as evidenced by the appearance of autophagic vacuoles and activation of autophagic marker proteins. Such GO-induced autophagy was observed in various cell lines and in macrophage treated with GO of different sizes. Strikingly, GO treatment of macrophage provoked the toll-like receptor (TLR) signaling cascades and triggered ensuing cytokine responses. Molecular analysis identified that TLR4 and TLR9 and their downstream signaling mediators MyD88, TRAF6 and NF-κB played pivotal roles in the GO-induced inflammatory responses. By silencing individual genes in the signaling pathway, we further unveiled that the GO-induced autophagy was modulated by TLR4, TLR9 and was dependent on downstream adaptor proteins MyD88, TRIF and TRAF6. Altogether, we demonstrated that GO treatment of cells simultaneously triggers autophagy and TLR4/TLR9-regulated inflammatory responses, and the autophagy was at least partly regulated by the TLRs pathway. This study thus suggests a mechanism by which cells respond to nanomaterials and underscores the importance of future safety evaluation of nanomaterials.

Introduction

Graphene and its oxidized form, graphene oxide (GO), have drawn intense attention in recent years for biological and medical applications [1]. The surface of GO contains hydrophilic oxygen-containing functional groups (i.e. hydroxyl, epoxyl and carboxyl tails) on the basal plane and edges, rendering GO amenable to stable dispersion in water and functionalization. These attributes have prompted the use of GO for bioimaging [2], cellular probing [3], cellular growth and differentiation [4], gene and drug delivery [5], [6] and photothermal therapy [7]. These burgeoning applications in biomedicine entail the need to evaluate the in vitro and in vivo safety of GO.

Autophagy is a process that degrades intracellular components in response to stressful conditions (e.g. starvation and infection) and is linked to cellular processes as diverse as cell survival, cell death, pathogen clearance and antigen presentation [8], [9], [10]. Autophagy involves the formation of double-membraned vesicles termed autophagosomes, which sequester cytoplasm and organelles and then fuse with lysosomes to form autolysosomes, thus degrading the contents of the vacuole [10]. Autophagy is negatively controlled by mTOR (mammalian target of rapamycin) complex 1 (mTORC1) and inhibition of mTORC1 kinase activity initiates the formation of autophagosome that comprises a complex consisting of Beclin 1 and other factors. The autophagosome formation also involves the conversion of microtubule-associated protein light chain 3 (LC3-I) to the lipidated form LC3-II, consequently conversion from LC3-I to LC3-II is a common indicator of autophagy.

Toll-like receptors (TLRs) are important receptors for the detection of microbial antigens and subsequent induction of innate immune responses [11]. Among the TLRs, TLR2 recognizes bacterial lipoproteins while TLR3 detects virus-derived dsRNA. TLR4 recognizes lipopolysaccharides (LPS) and TLR5 recognizes bacterial flagellin. TLR7 mediates recognition of viral ssRNA while TLR9 senses unmethylated DNA with CpG motifs derived from bacterial and viruses [12]. Upon engagement with cognate ligands, the TLRs transduce signals by first recruiting adaptor proteins including myeloid differentiating factor 88 (MyD88) and TIR domain-containing adaptor inducing IFN-beta (TRIF), followed by activation of downstream signaling proteins such as TRAF6 and NF-κB, eventually resulting in various cellular responses including secretion of cytokines and interferons (IFNs).

The connection between autophagy and TLRs was discovered in 2007 as it was found that TLRs signaling in macrophages links the autophagy pathway to phagocytosis [13] and TLR4 stimulation enhances the autophagic elimination of phagocytosed mycobacteria in macrophages [14]. Ensuing studies further reported that TLR2, TLR3 and TLR7 play roles in autophagy induction [15], [16]. The TLR-activated autophagy is regulated by the interaction of MyD88 or TRIF with Beclin 1 [17] and TLR engagement induces the interaction, thereby reducing the binding of Beclin 1 to inhibitory molecules. Beclin 1 also interacts with TRAF6 so as to facilitate its activation and subsequent formation of autophagosomes [18], thus TRAF6 seems to be crucial for TLR-activated autophagy [18]. The regulatory interaction of heat shock protein HSP90 with Beclin 1 is also required for autophagy induction downstream of TLR3 and TLR4 [19].

Since GO holds promise for diverse in vivo applications and macrophage is the primary immune cell type that engages foreign substances and modulates the immune responses in vivo, the overriding objective of this study was to explore how macrophage responded to GO treatment in order to evaluate the safety of GO.

Section snippets

Preparation and characterization of GO

Large GO with a size of ≈2.4 μm was prepared from natural graphite (Bay Carbon, SP-1, average particle size ≈30 μm) by the modified Hummers method as described previously [20] and dispersed in water. The solution was centrifuged (7200 × g for 5 min) to remove unexfoliated GO and byproducts and centrifuged again (400 × g for 15 min) to remove broken fragments and debris. The pellet was dried under vacuum overnight to yield the large GO, weighed on a Sartorius SE2 ultra-micro balance with 0.1 μg

Preparation and characterization of large and small GO nanosheets

Fig. 1A schematically illustrates the preparation of GO nanosheets, for which large-size GO was prepared from natural graphite by the modified Hummers method as described [20] while small-size GO was obtained by sonicating large GO into smaller pieces via tip sonication. Atomic force microscopy (AFM) images showed significant difference of lateral dimensions between large and small GO (Fig. 1B–C). The thicknesses of both large and small GO measured ≈1.0–1.2 nm, which agreed with the GO

Discussion

Recent publications have shown that nanomaterials such as quantum dots [26], polymer dendrimers [29], fullerene [30], gold nanoparticles [31], carbon nanotube [32] and αAl2O3 nanoparticles [25] can induce autophagy. However, the underlying mechanisms contributing to the nanomaterials-induced autophagy remain elusive. Very recently, GO nanosheets with different lateral dimensions (≈2 μm and 350 nm) were found to be internalized by macrophages via phagocytosis, resulting in cell death and

Conclusions

In summary, this study demonstrated that treatment of cells with GO simultaneously triggers autophagy and TLR4/TLR9-regulated inflammatory responses, and the autophagy was at least partly regulated by the TLRs pathway. Since TLRs-regulated autophagy can contribute to efficient autophagic elimination of intracellular microbes such as Bacillus Calmette-Guerin and Listeria monocytogenes, it is likely that GO-triggered autophagy is exploited by the cells to clear the internalized GO. However, this

Acknowledgment

The authors acknowledge the financial support from the National Tsing Hua University (Booster Program 99N2544E1 and Toward World-Class University Project 100N2050E1, 101N2060E1) and National Science Council (99-2221-E-007-025-MY3, 100-2628-E-007-029-MY2, 100-2325-B-080-001), Taiwan.

References (35)

  • J.J. Li et al.

    Autophagy and oxidative stress associated with gold nanoparticles

    Biomaterials

    (2010)
  • H. Yue et al.

    The role of the lateral dimension of graphene oxide in the regulation of cellular responses

    Biomaterials

    (2012)
  • S. Akira et al.

    Pathogen recognition and innate immunity

    Cell

    (2006)
  • Q. Liu et al.

    Graphene and graphene oxide sheets supported on silica as versatile and high-performance adsorbents for solid-phase extraction

    Angew Chem Int Ed Engl

    (2011)
  • H. Kim et al.

    Graphene oxide-polyethylenimine nanoconstruct as a gene delivery vector and bioimaging tool

    Bioconjug Chem

    (2011)
  • Y. Wang et al.

    Aptamer/graphene oxide nanocomplex for in situ molecular probing in living cells

    J Am Chem Soc

    (2010)
  • L. Zhang et al.

    Enhanced chemotherapy efficacy by sequential delivery of siRNA and anticancer drugs using PEI-grafted graphene oxide

    Small

    (2011)
  • Cited by (214)

    • Graphene oxide as novel vaccine adjuvant

      2023, International Immunopharmacology
    View all citing articles on Scopus
    View full text