Elsevier

Journal of Controlled Release

Volume 117, Issue 2, 12 February 2007, Pages 148-162
Journal of Controlled Release

Review
The taming of the cell penetrating domain of the HIV Tat: Myths and realities

https://doi.org/10.1016/j.jconrel.2006.10.031Get rights and content

Abstract

Protein transduction with cell penetrating peptides over the past several years has been shown to be an effective way of delivering proteins in vitro and now several reports have also shown valuable in vivo applications in correcting disease states. An impressive bioinspired phenomenon of crossing biological barriers came from HIV transactivator Tat protein. Specifically, the protein transduction domain of HIV Tat has been shown to be a potent pleiotropic peptide in protein delivery. Various approaches such as molecular modeling, arginine guanidinium head group structural strategy, multimerization of PTD sequence and phage display system have been applied for taming of the PTD. This has resulted in identification of PTD variants which are efficient in cell membrane penetration and cytoplasmic delivery. In spite of these state of the art technologies, the dilemma of low protein transduction efficiency and target specific delivery of PTD fusion proteins remains unsolved. Moreover, some misconceptions about PTD of Tat in the literature require considerations. We have assembled critical information on secretory, plasma membrane penetration and transcellular properties of Tat and PTD using molecular analysis and available experimental evidences.

Introduction

The past decade has witnessed tremendous advances in the field of protein transduction, aiming to correct defects for proteins involved in a variety of disease processes. At present, it is possible to produce a given protein molecule by recombinant DNA technology for in vivo therapeutic applications. Nevertheless, it still remains a challenge to deliver the recombinant proteins to desired targets in vivo, although small molecules or peptides capable of crossing cellular membranes have been successfully designed to deliver small or moderately large proteins. Despite developments in the area of protein transduction peptides, the classical delivery methods of protein coding genes via adeno-associated virus (AAV) [1], [2], adenovirus (AV) [3], [4], lentivirus [5], herpes virus (HSV) [6], [7] vectors, and plasmid expression vectors [8], [9] remain the preferred choice for expression of proteins.

Because of their natural abilities in delivering the specific genes to permissive cells, viral vector-mediated gene expression is considered the most efficient and reliable approach for expressing functional proteins de novo in mitotically active or post-mitotically blocked cell types (HIV viral vectors). Nonetheless, viral vectors invariably are required in large doses to achieve therapeutic expression levels of intended protein (s). Moreover, viral vectors integrate with the host chromatin material. These properties may have consequences from long term effects on host genetic systems, and therefore, safety remains a serious concern for their ultimate clinical application [10], [11], [12], [13].

An alternative approach that appears to be the safest is to produce recombinant proteins exogenously and then deliver them systemically or by localized injections into the target organs. The delivery and bioavailability of recombinant proteins into cells or tissues need further improvements, however. Discovery of the HIV Tat protein transduction domain (PTD) has opened avenues for directing in vitro and in vivo delivery of proteins into cells. Several studies have shown the potential of PTD in drug delivery [14], [15] and transduction of proteins as large as 110 kDa into different cells [16]. In vivo injection of fusion proteins systemically has demonstrated the effectiveness of the PTD in protein delivery [15], [16]. In the present review we discuss the current status of the protein transduction focusing mainly on the PTD domain of HIV Tat.

Section snippets

Cell penetrating peptides (CPPs) for protein delivery

Various approaches have been designed to develop CPPs for introducing recombinant proteins into the cells. Penetratin [17], polylysine [18], [19], polyarginine [20], Tat-PTD [16], [21], HSV VP22 [22], [23], [24], Kaposi FGF [25], Syn B1 [26], FGF-4 [27], [28], nuclear localization signal (NLS) [29], and anthrax toxin derivative 254-amino acids (aa) peptide segment [30], diphtheria toxin ‘R’ binding domain [31], MPG (HIV gp41/SV40 Tag NLS) [32], pep-1 [33], WR peptide [34], and exotoxin A [35]

Potential of PTD fusion protein transduction in vitro

Several years ago, Frankel and Pabo [48] and Green and Lowenstein [49] demonstrated that extracellular HIV Tat can cross the plasma membrane and enter the cell, reaching the nucleus. The successful entry of extracellular Tat was investigated by others and has become a common test to monitor HIV-LTR promoter activity. Subsequently, more detailed analysis of Tat protein [21], [47], [50], [51] identified a PTD of 9–11 aa residues, a basic domain that can transduce itself, as well as the bigger

Potential of PTD fusion protein transduction in vivo

In 1999 Schwarze et al. [16] showed that PTD-beta-gal fusion protein applied intraperitoneally entered the brain after crossing the blood–brain barrier (BBB). Subsequently, several other studies have also reported effective biodistribution of therapeutically important proteins in vivo by using PTD [52], [69], [70], [71], [72], [73], [74], [75]. In a previous study, however, no PTD-mediated transduction in the brain was observed [46], possibly because beta-gal was chemically conjugated to PTD

Mechanism of PTD internalization

It has been observed that histones and cationic polyamines such as polylysine stimulate the uptake of albumin by tumor cells in culture [85], [86]. But, the cellular uptake mechanism(s) of CPPs is currently unknown. CPPs are structurally diverse and highly variable in nature. Nonetheless, their common feature is the high density of basic amino acid residues (Arg and Lys): the presence of basic amino acids in the PTD is considered the hallmark of transduction peptides. There are exceptions,

Role of lysosomotropic agents in PTD-mediated protein transduction

Some studies have reported inefficient delivery of PTD-based fusion proteins in vitro [66], [113], [133]. The failure of PTD-mediated protein transduction was explained in two ways. First, nuclear translocation of fusion proteins deposited on the plasma membrane after fixation of cells has been the common explanation, and hence PTD actually does not transduce proteins. Second, deposits of PTD fusion proteins on the cell surface with no biological activity were demonstrated. These studies may

Transcellular property of PTD

Successful protein transductions have resulted in the investigation of the transcellular effect on bystander cells. Limited studies have shown that PTD is conferred with transcellular property [125], [141]. Indeed, this behavior is extremely valuable in delivering therapeutic proteins to surrounding cells. Therefore, it is important to know whether a preformed PTD fusion protein or a DNA expression vector for the PTD fusion protein is involved in transcellular transduction. Of note, Tat-PTD is

Validation of true PTD-mediated protein transduction

PTDs are promising tools for transducing presynthesized proteins across the plasma membrane. Nonetheless, because artifacts result from fixation and endosomal entrapment, true cytosolic distribution or targeting to nucleus is hampered by the use of nonvisual methods [134]. There are limited numbers of approaches that can verify the true nature of PTD fusion protein transduction in vitro. The one most commonly employed is trypsin treatment, which removes the surface-bound fusion proteins and

Cytotoxicity of PTD

There are limited toxicity studies on PTD peptide. Tat peptide of 48–85 aa encompassing a PTD domain did not show neurotoxicity in cultures in vitro [166], but prolonged exposure (24 h) of Hela cells to Tat peptide containing alpha helical sequence (37–60) resulted in necrosis of 60% of cells [47] while Tat peptide (43–60) revealed 10–15% toxicity. Short exposure of cell cultures with 20–100 μM concentrations of PTD did not exhibit any adverse effects [47], [167], whereas a 500 μM dose of PTD

Limitations in PTD-mediated protein transduction

Since the inception of protein delivery studies, cell-to-cell movement of transduced fusion protein has not been thoroughly studied. Furthermore, in reality, PTD-based delivery of fusion proteins will invariably result in nuclear targeting [63], [135], [142], [143], [144], [145], [146], [147], [148], [168] which may not be required in every case. It is useful, however, in some cases such as expression of single chain antibody fragment, where the product is required in the nucleus to inhibit

Challenges and future of PTD

PTD fusion proteins have great potential, especially for in vitro studies. Application of PTD-directed protein delivery in vivo could prove useful under certain situations, where immediate administration of presynthesized proteins is required. But because of limited evidence and lack of a uniform means of producing proteins, and the poor protein transduction property of PTD, its wide scale use is likely to be delayed. Furthermore, because of inherent variations in the properties of different

Conclusions

PTD of HIV Tat protein is a nuclear localization signal which is conferred with mild protein transduction property. PTD delivers the cargo to the nucleus, but lacks transcellular property. The taming of the PTD has resulted in more potent PTDs for cytoplasmic delivery. Finally, PTD may be envisioned as a universal protein and nucleic acid transducer but obviously not in the present form.

Acknowledgements

The work is supported by NIH grant (RO1 NS050064) to AC. AC also thanks Drs. Krister Kristensson, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden; Suzanne Gartner, Avindra Nath and Pamela Talalay, Department of Neurology, Johns Hopkins University, Baltimore, USA, for discussions and facilities.

References (183)

  • G.P. Dietz et al.

    Inhibition of neuronal apoptosis in vitro and in vivo using TAT-mediated protein transduction

    Mol. Cell. Neurosci.

    (2002)
  • D. Klein et al.

    Delivery of Bcl-XL or its BH4 domain by protein transduction inhibits apoptosis in human islets

    Biochem. Biophys. Res. Commun.

    (2004)
  • W. Wang et al.

    Targeting p53 by PTD-mediated transduction

    Trends Biotechnol.

    (2004)
  • D. Ozaki et al.

    Transduction of anti-apoptotic proteins into chondrocytes in cartilage slice culture

    Biochem. Biophys. Res. Commun.

    (2004)
  • Y.D. Kwon et al.

    Cellular manipulation of human embryonic stem cells by TAT-PDX1 protein transduction

    Molec. Ther.

    (2005)
  • D.M. Andrade et al.

    Protein therapy for Unverricht–Lundborg disease using cystatin-B transduction by Tat-PTD. Is it that simple

    Epilepsy Res.

    (2006)
  • M. Lundberg et al.

    Positively charged DNA-binding proteins cause apparent cell membrane translocation

    Biochem. Biophys. Res. Commun.

    (2002)
  • I. Tikhonov et al.

    Furin cleavage of the HIV-1 Tat protein

    FEBS Lett.

    (2004)
  • S.S. Elliger et al.

    Enhanced secretion and uptake of beta-glucuronidase improves adeno-associated viral-mediated gene therapy of mucopolysaccharidosis type VII mice

    Molec. Ther.

    (2002)
  • S. Krautwald et al.

    Transduction of the TAT-FLIP fusion protein results in transient resistance to Fas-induced apoptosis in vivo

    J. Biol. Chem.

    (2004)
  • D. Kim et al.

    Cytoplasmic transduction peptide (CTP): new approach for the delivery of biomolecules into cytoplasm in vitro and in vivo

    Exp. Cell Res.

    (2006)
  • S.R. Cai et al.

    The kinetics and tissue distribution of protein transduction in mice

    Eur. J. Pharm. Sci.

    (2006)
  • J.S. Wadia et al.

    Protein transduction technology

    Curr. Opin. Biotechnol.

    (2002)
  • T. Niidome et al.

    Binding of cationic alpha-helical peptides to plasmid DNA and their gene transfer abilities into cells

    J. Biol. Chem.

    (1997)
  • A. Ziegler et al.

    Interaction of the protein transduction domain of HIV-1 TAT with heparan sulfate: binding mechanism and thermodynamic parameters

    Biophys. J.

    (2004)
  • S. Sandgren et al.

    Nuclear targeting of macromolecular polyanions by an HIV-Tat derived peptide. Role for cell-surface proteoglycans

    J. Biol. Chem.

    (2002)
  • M. Tyagi et al.

    Internalization of HIV-1 tat requires cell surface heparan sulfate proteoglycans

    J. Biol. Chem.

    (2001)
  • M. Rusnati et al.

    Interaction of HIV-1 Tat protein with heparin. Role of the backbone structure, sulfation, and size

    J. Biol. Chem.

    (1997)
  • M. Yanagishita et al.

    Cell surface heparan sulfate proteoglycans

    J. Biol. Chem.

    (1992)
  • S. Console et al.

    Antennapedia and HIV transactivator of transcription (TAT) “protein transduction domains” promote endocytosis of high molecular weight cargo upon binding to cell surface glycosaminoglycans

    J. Biol. Chem.

    (2003)
  • A.K. Hennig et al.

    Intravitreal gene therapy reduces lysosomal storage in specific areas of the CNS in mucopolysaccharidosis VII mice

    J. Neurosci.

    (2003)
  • K. Shimazaki et al.

    Adeno-associated virus vector-mediated bcl-2 gene transfer into post-ischemic gerbil brain in vivo: prospects for gene therapy of ischemia-induced neuronal death

    Gene Ther.

    (2000)
  • M. Akimoto et al.

    Adenovirally expressed basic fibroblast growth factor rescues photoreceptor cells in RCS rats

    Invest. Ophthalmol. Visual Sci.

    (1999)
  • S. Kugler et al.

    Human synapsin 1 gene promoter confers highly neuron specific long-term transgene expression from an adenoviral vector in the adult rat brain depending on the transduced area

    Gene Ther.

    (2003)
  • J.H. Kordower et al.

    Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease

    Science

    (2000)
  • M.S. Lawrence et al.

    Herpes simplex viral vectors expressing Bcl-2 are neuroprotective when delivered after a stroke

    J. Cereb. Blood Flow Metab.

    (1999)
  • M.A. Hickman et al.

    Gene expression following direct injection of DNA into liver

    Hum. Gene Ther.

    (1994)
  • U.R. Hengge et al.

    Expression of naked DNA in human, pig, and mouse skin

    J. Clin. Invest.

    (1996)
  • E. Check

    Gene therapy: shining hopes dented—but not dashed

    Nature

    (2002)
  • J.B. Connolly

    Lentiviruses in gene therapy clinical research

    Gene Ther.

    (2002)
  • K. Cornetta et al.

    Safety issues related to retroviral-mediated gene transfer in humans

    Hum. Gene Ther.

    (1991)
  • E. Marshall

    Viral threat to newborns under radar

    Science

    (2002)
  • B. Gupta et al.

    Transactivating transcriptional activator-mediated drug delivery

    Expert Opin. Drug Deliv.

    (2006)
  • S.R. Schwarze et al.

    In vivo protein transduction: delivery of a biologically active protein into the mouse

    Science

    (1999)
  • W.C. Shen et al.

    Conjugation of poly-l-lysine to albumin and horseradish peroxidase: a novel method of enhancing the cellular uptake of proteins

    Proc. Natl. Acad. Sci. U. S. A.

    (1978)
  • H.J. Ryser et al.

    The cellular uptake of horseradish peroxidase and its poly (lysine) conjugate by cultured fibroblasts is qualitatively similar despite a 900-fold difference in rate

    J. Cell. Physiol.

    (1982)
  • D.J. Mitchell et al.

    Polyarginine enters cells more efficiently than other polycationic homopolymers

    J. Pept. Res.

    (2000)
  • H. Nagahara et al.

    Transduction of full-length TAT fusion proteins into mammalian cells: TAT-p27Kip1 induces cell migration

    Nat. Med.

    (1998)
  • M.S. Dilber et al.

    Intercellular delivery of thymidine kinase prodrug activating enzyme by the herpes simplex virus protein, VP22

    Gene Ther.

    (1999)
  • C.S. Liu et al.

    VP22 enhanced intercellular trafficking of HSV thymidine kinase reduced the level of ganciclovir needed to cause suicide cell death

    J. Gene Med.

    (2001)
  • Cited by (147)

    • Control of solvent exposure of cationic polypeptides in anionic environment

      2020, Chemical Physics Letters
      Citation Excerpt :

      Thus, the charged groups in proteins are more flexible compared to rigid backbone of a DNA molecule. Secondly, counter-anion condensation is favoured by helical conformational property of the peptide backbone [19–22]. The cationic domains in CPPs are rich in arginine, a hydrophilic amino acid residue with positively charged basic guanidinium side chain at normal pH [29].

    View all citing articles on Scopus
    View full text