Elsevier

Journal of Controlled Release

Volume 147, Issue 2, 15 October 2010, Pages 171-179
Journal of Controlled Release

Cationic cell-penetrating peptides induce ceramide formation via acid sphingomyelinase: Implications for uptake

https://doi.org/10.1016/j.jconrel.2010.06.030Get rights and content

Abstract

Cationic cell-penetrating peptides (CPP) are receiving increasing attention as molecular transporters of membrane-impermeable molecules. Import of cationic CPP occurs both via endocytosis and — at higher peptide concentrations — in an endocytosis-independent manner via localized regions of the plasma membrane. At present, this endocytosis-independent import of cationic CPP is not well understood, but has been shown to be sensitive to various pharmacological inhibitors, suggesting a role of an unidentified enzymatic activity. Here, we demonstrate that the direct translocation of cationic CPP depends on a CPP-induced translocation of acid sphingomyelinase (ASMase) to the outer leaflet of the plasma membrane and ceramide formation. The involvement of ASMase in uptake was confirmed by a pharmacological inhibition of ASMase by imipramine and a subsequent rescue of uptake through external addition of sphingomyelinase, and by using ASMase-deficient cells. We also found that the threshold for direct CPP translocation can be lowered through addition of sphingomyelinase and that sphingomyelinase enhances the translocation of R9 coupled to low-molecular weight cargos, but not high-molecular weight cargos. In conclusion, we show that a previously poorly understood mechanism of cationic CPP import depends on the ASMase-dependent formation of ceramide on the outer leaflet of the plasma membrane. To our knowledge, this is the first illustration that a class of delivery vectors operates through the induction of an enzymatic activity that changes the lipid composition of the plasma membrane.

Introduction

Interest in cell-penetrating peptides (CPP) as tools to deliver membrane-impermeable therapeutic molecules continues to grow. This development is illustrated by the increasing efforts of the scientific community to elucidate the chemical, physical and biological principles underlying the activity of CPP and CPP-based delivery systems [1], [2], [3], [4] and the steep rise in the number of therapeutic strategies that are being pursued with these systems [5], [6]. It is becoming progressively clear that instead of a single mechanism that is valid for all CPP, multiple modes-of-action exist with respect to the route of internalization and intracellular trafficking, that depend on the cell line [7], the CPP [8], and the cargo [9], [10]. Although endocytosis is now considered the major internalization route of cationic CPP [4], [11], [12], [13], [14], direct translocation for cationic CPP has also been observed, mainly at high [8], [15], [16], [17], but also at low concentrations [18], [19].

For arginine-rich CPP the potential for direct translocation is thought be related to the ability of the guanidinium moieties of arginines to form bidentate hydrogen bonds with membrane lipids [20] and is underscored by the ability of the arginine-rich TAT peptide to induce pores in artificial membranes, as was observed by small angle X-ray scattering [21]. Moreover, molecular-dynamics simulations indicate the potential of arginine-rich peptides to form transient pores in membranes in the presence of an electrochemical gradient and provide a mechanistic hypothesis for direct membrane translocation [22].

Nevertheless, biological details remain poorly understood and it is not clear to which degree in vitro models manage to reflect the complex molecular environment of a cell. At higher concentrations, direct cytoplasmic import of cationic CPP has been shown to depend on spatially confined nucleation zones. This rapid import is sensitive to the PKCδ inhibitor rottlerin and to chlorpromazine [8]. Available evidence indicates that the plasma membrane remains intact.

In a search for a common denominator of the observed pharmacological profile, we discovered that both molecules act as inhibitors of acid sphingomyelinase (ASMase) activity [23], [24], [25]. This finding provided us with a rationale for investigating a possible involvement of ASMase in the CPP uptake via direct translocation.

Acid sphingomyelinase has traditionally been known in relation to Niemann–Pick disease, a lysosomal storage disorder [26]. More recently, an important role for this enzyme in ceramide-mediated signal transduction pathways has emerged, linking its activity to a variety of common diseases, among which are cancer, cardiovascular diseases and diabetes [27]. A major function of this enzyme is the hydrolysis of sphingomyelin to ceramide and phosphorylcholine following a trigger-initiated translocation from lysosomes to the outer leaflet of the plasma membrane. The details of the translocation mechanism are unknown at present. Ceramide is a lipid with a well-established role in the induction of antiproliferative and apoptotic responses in a variety of cancer cells [28]. As a part of this role, ceramide greatly affects the structure and properties of cellular membranes. In the plasma membrane ceramide-enriched membrane domains modulate signaling [29], [30]. In the mitochondrial outer membrane ceramide increases permeability [31]. Here, we provide evidence for a role of ceramide formation by ASMase at the plasma membrane in the rapid cytoplasmic import of cationic CPP. This finding gives rise to the concept that cationic CPP enhance their own uptake by initiating a positive feed-back loop that involves an enzymatic alteration of the lipid composition of the plasma membrane.

Section snippets

Materials

C-terminally amidated peptides were purchased from EMC microcollections (Tübingen, Germany). Fluorophore-labeled peptides were synthesized with an N-terminal carboxyfluorescein-label. Purity was evaluated by high-performance liquid chromatography (HPLC) and identity confirmed by mass spectrometry. If required, peptides were further purified to a purity of > 95% using reversed-phase HPLC. The CPP hLF was oxidized before use as described previously [32]. Bovine serum albumin (BSA), glucose,

Inhibition of direct translocation of nona-arginine (R9) after inhibition of ASMase

Previously, we and others described the direct cytoplasmic import of cationic CPP through localized plasma membrane regions, which we had termed nucleation zones [8], [15], [17]. This import has a particular pharmacological profile, being sensitive to rottlerin, and chlorpromazine. In a search for a common denominator of these findings, we found that both compounds have the potential to interfere with the activation of ASMase. Among other activities, rottlerin has been shown to directly prevent

Discussion

CPP hold a great potential for the delivery of membrane-impermeable molecules. To exploit this potential to the fullest, it is imperative that the import mechanisms and the associated effects on the cells are well understood. Despite various reports of direct translocation of arginine-rich CPP [8], [15], [16], [17], [20], and biophysical [21] and molecular dynamics [22] studies supporting this ability, the biology behind these observations has proven elusive. Here, we demonstrate that the

Competing interests

The authors declare no competing financial interests.

Acknowledgements

We thank F. Valsecchi (Radboud University Nijmegen Medical Centre) for providing primary mouse feet fibroblasts. The authors acknowledge financial support from the Volkswagen-Foundation (Nachwuchsgruppen an Universitäten, I/77 472) and from the Radboud University Nijmegen Medical Centre to WPRV. Melissa Thanos was supported by the IFORES program. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References (60)

  • F. Duchardt et al.

    A cell-penetrating peptide derived from human lactoferrin with conformation-dependent uptake efficiency

    J. Biol. Chem.

    (2009)
  • J.P. Richard et al.

    Cell-penetrating peptides. A reevaluation of the mechanism of cellular uptake

    J. Biol. Chem.

    (2003)
  • D. Delaroche et al.

    Cell-penetrating peptides with intracellular actin-remodeling activity in malignant fibroblasts

    J. Biol. Chem.

    (2010)
  • C. Du et al.

    Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition

    Cell

    (2000)
  • H. Grassme et al.

    Rhinoviruses infect human epithelial cells via ceramide-enriched membrane platforms

    J. Biol. Chem.

    (2005)
  • F.M. Goni et al.

    Sphingomyelinases: enzymology and membrane activity

    FEBS Lett.

    (2002)
  • J.A. Rotolo et al.

    Caspase-dependent and -independent activation of acid sphingomyelinase signaling

    J. Biol. Chem.

    (2005)
  • H. Qiu et al.

    Activation of human acid sphingomyelinase through modification or deletion of C-terminal cysteine

    J. Biol. Chem.

    (2003)
  • J.H. Kuo et al.

    Interactions between octaarginine and U-937 human macrophages: global gene expression profiling, superoxide anion content, and cytokine production

    J. Control. Release

    (2009)
  • M.B. Ruiz-Arguello et al.

    Different effects of enzyme-generated ceramides and diacylglycerols in phospholipid membrane fusion and leakage

    J. Biol. Chem.

    (1996)
  • L.C. Silva et al.

    Lipid raft composition modulates sphingomyelinase activity and ceramide-induced membrane physical alterations

    Biophys. J.

    (2009)
  • R.W. Jenkins et al.

    Roles and regulation of secretory and lysosomal acid sphingomyelinase

    Cell. Signal.

    (2009)
  • N.T. Xuan et al.

    Sphingomyelinase dependent apoptosis of dendritic cells following treatment with amyloid peptides

    J. Neuroimmunol.

    (2010)
  • M. Magzoub et al.

    Erratum: Cell-penetrating peptides: From inception to application

    Q. Rev. Biophys.

    (2004)
  • M.C. Morris et al.

    Cell-penetrating peptides: from molecular mechanisms to therapeutics

    Biol. Cell

    (2008)
  • L.N. Patel et al.

    Cell penetrating peptides: intracellular pathways and pharmaceutical perspectives

    Pharm. Res.

    (2007)
  • R. Fischer et al.

    Break on through to the other side-biophysics and cell biology shed light on cell-penetrating peptides

    Chembiochem

    (2005)
  • K.M. Stewart et al.

    Cell-penetrating peptides as delivery vehicles for biology and medicine

    Org. Biomol. Chem.

    (2008)
  • J. Mueller et al.

    Comparison of cellular uptake using 22 CPPs in 4 different cell lines

    Bioconjug. Chem.

    (2008)
  • F. Duchardt et al.

    A comprehensive model for the cellular uptake of cationic cell-penetrating peptides

    Traffic

    (2007)
  • Cited by (77)

    • Ionpair-π interactions favor cell penetration of arginine/tryptophan-rich cell-penetrating peptides

      2020, Biochimica et Biophysica Acta - Biomembranes
      Citation Excerpt :

      These interactions appear to be critical for GAG-dependent internalization. For direct translocation, our DSC data suggest that peptides containing 2 or 3 Trp promote lateral phase segregation, and this type of translocation mechanism has previously been proposed for CPPs [34,42,51]. Finally, with 4 Trp, internalization is greatly enhanced.

    • Membrane permeation of arginine-rich cell-penetrating peptides independent of transmembrane potential as a function of lipid composition and membrane fluidity

      2017, Journal of Controlled Release
      Citation Excerpt :

      Remarkably, while the plasma membrane of HeLa cells was rich in sphingomyelin, hardly any sphingomyelin was detected in the isolates of HEK cells (Fig. 2). When HeLa cells were treated with bSMase, ceramide could be detected, which is in agreement with immunofluorescence staining in a previous study that showed colocalization of NZ with ceramide [9]. The results so far indicated that the plasma membrane of HeLa cells is resistant to peptide penetration, and direct uptake can occur only after modulation of membrane composition through sphingomyelin turnover.

    View all citing articles on Scopus
    View full text