Skip to main content

Biological activities of histidine-rich peptides; merging biotechnology and nanomedicine

Abstract

Histidine-rich peptides are commonly used in recombinant protein production as purification tags, allowing the one-step affinity separation of the His-tagged proteins from the extracellular media or cell extracts. Genetic engineering makes feasible the post-purification His-tag removal by inserting, between the tag and the main protein body, a target site for trans-acting proteases or a self-proteolytic peptide with regulatable activities. However, for technical ease, His tags are often not removed and the fusion proteins eventually used in this form. In this commentary, we revise the powerful biological properties of histidine-rich peptides as endosomolytic agents and as architectonic tags in nanoparticle formation, for which they are exploited in drug delivery and other nanomedical applications. These activities, generally unknown to biotechnologists, can unwillingly modulate the functionality and biotechnological performance of recombinant proteins in which they remain trivially attached.

Affinity protein purification

Protein engineering and production, as a widely spread methodological platform, is providing reagents for catalysis, drugs for pharmaceutical industries and in general, instruments for analytical research in structural biology, proteomics, interactomics and drug design, among others. If secreted, recombinant proteins produced in prokaryotic or eukaryotic cell factories are found diluted in the medium, or alternatively, they occur as minor components of extremely complex macromolecular mixtures if retained inside the cell. Therefore, they must be purified so as achieve conveniently high concentration and purity levels required for stable storage and characterization or for efficient use in biological systems or in catalysis. The identification of peptide tags for single-step affinity-based purification, upon end terminal fusion, has provided an advantageous tool that represents a faster, easy and cost-effective alternative to multiple-step chromatographic separations [1, 2]. Protein tagging allows the purification of virtually any protein without previous knowledge of its properties or those of its expected contaminants, since the capturing event relies on tag's rather than on protein's features.

Being absent in the natural target protein, affinity tags are expected to be removed upon purification, to keep the target protein sequence as natural as possible [3, 4]. During the upstream cloning strategies, recombinant proteins can be designed to contain target sites for trans-acting proteases [4] or alternatively, functional endoproteolytic peptides [5] to remove the additional peptides. The Tobacco Etch Virus TEV protease (TEVp) is probably the most used agent for trans tag removal, because of its high specificity for the TEVp target site (ENLYFQ↓G) compared to the most promiscuous behaviour of alternative enzymes such as factor Xa, enterokinase and some picornaviridae 3C proteases [4]. On the other hand, inteins are among the most used cis-acting tag-removal agents [6–8].

However, for technical convenience, the foreign peptide is often not removed after purification, as for instance, the C-position of the cleavage site in the recognition sequence of most endoproteases makes specially difficult the complete removal of C-terminal added stretches [1]. Obviously, the modification of the primary amino acid sequence is expected to have effects on physicochemical parameters of the protein such as the distribution of charged amino acids and the final conformation, and also on biological properties such as in intra- or intermolecular interactions, solubility and enzymatic activity. However, some observations suggest that such effects might be extremely soft or even convenient. In this context, a few additional end terminal amino acids seem not affecting the packing of the crystals or atomic interactions, leading to the correct 3D structure determination [9]. Furthermore, the presence of purification tags might positively affect the solubility, productivity and proteolytic stability of recombinant proteins [10–14], which would provide an added value to these protein segments. These observations have prompted researchers to evaluate affinity tags not only for their usefulness as tools for downstream but also as midstream folding-assistant agents [1]. Finally, some analytical procedures to determine in vivo protein-protein interactions (such as the tandem affinity purification) are based on the presence in the model protein of affinity purification tags [15, 16], relying on the assumption that these additional segments do not dramatically influence the protein's natural capability to perform cross-molecular interactions.

Histidine-rich peptides in Biotechnology

Histidine-rich peptides (often H6 but also other peptide versions, Table 1) are probably the most used protein purification tags [1]. Being short sequences, His-tags do not add significant metabolic load to the protein production process and they can be easily incorporated to the protein by simple genetic engineering at the upstream level. Since the interaction between a His-tag and its ligand does not require any specific conformation, binding is feasible under either native or denaturing conditions (e.g. urea 8 M or guanidinium hydrochloride 6 M) [17] as well as under oxidizing or reducing conditions, resulting in high technological flexibility and easy scalability [18].

Table 1 Representative examples of His-rich peptides used in biotechnology as affinity tags for protein purification.

Immobilized metal affinity chromatography (IMAC) [19] is a separation principle based on the differential and reversible affinity of transition metal ions such as Zn2+, Cu2+, Ni2+, and Co2+ for histidines [20], due to the coordination bonds formed between metal ions and amino acid side chains exposed on the protein surface. Electron-donor atoms (N, S, O) present in the chelating compounds of the chromatographic support are capable of coordinating metal ions and forming metal chelates, which can be bidentate, tridentate, etc., depending on the number of occupied coordination bonds [21]. Four of the six coordination sites on the octahedral Ni2+ center are occupied by the nitrilotriacetic acid (NTA) ligand, and the remaining coordination sites are occupied by two of the six imidazole moieties in the H6-tag. The model in which the interaction of the metal ion with His-tag locates in residues n and n+2 of the tag is confirmed by the fact that IMAC ligands can bind to His-tags consisting of consecutive as well as to alternating histidine residues.

As the chelating ligand is used to fix the metal to agarose, iminodiacetic acid (IDA) was initially employed and it is still in use today in many commercial IMAC resins. This technology was improved by the invention of the chelating ligand NTA in the 1980s [22].

The amino acid histidine shows the strongest interaction with immobilized metal ion matrices, as electron donor groups on histidine imidazole ring readily form coordination bonds with the immobilized metal. In Biacore experiments, the dissociation rate of a H6-tagged protein to Ni-NTA was estimated to be 1 × 10-6 to 1.4 × 10-8 M at pH 7.0-7.4. The same study also showed that two histidines separated by either one or four other residues, are the preferred binding motifs [23]. Interestingly, selection of an optimum tag by a phage-displayed library showed that tags with only two histidine residues possessed chromatographic characteristics superior to those of the most commonly used H6-tag [24]. Adsorption of a protein to the IMAC support is usually performed in neutral or slightly basic media in which imidazole nitrogens in histidine residues are non-protonated. Elution of the target protein is achieved by protonation, ligand exchange or extraction of the metal ion by a stronger chelator, like EDTA.

Histidine-rich peptides in Nanomedicine

In a very different context, histidine-rich peptides are largely exploited in drug delivery as powerful endosomal escape agents, by their incorporation into drug-loaded nanoparticles or polyplexes to be internalized by target cells through endosome formation. During centripetal intracellular trafficking, most of the uptake routes converge into endocytic vesicles, where both vehicle and cargo may become enzymatically degraded when late endosomes are transformed into lysosomes [25]. Within endosomes, the imidazole group of histidine, with a pK around 6.0, gets protonated under the increasingly acid conditions. This step recruits Cl- ions with a consequent osmotic swelling and endosomal cracking (the 'proton sponge' model [26]), which results in the cytoplasmic release of the nanoparticle and escape from lysosomal enzymes. Histidine-rich peptides have been incorporated into polymers, liposomes and proteins (including virus-like particles, VLPs) (Table 2). In non-viral gene therapy, such histidine-empowered vehicles show up to almost 104-fold transgene expression improvement when compared with their corresponding histidine-lacking, parental versions [27]. Despite other membrane-active peptides, poly-histidines remain neutrally charged at neutral pH, avoiding non specific binding to serum proteins and consequent inactivation of the particle [28].

Table 2 Representative examples of His-rich peptides used in nanomedicine as endosomolytic agents.

The number of histidines and their distribution in the amino acid sequence of the tag determine endosomal escape efficiency of His-tagged vehicles and nanoparticles. Lo and Wang [27] showed that the endosomolytic activity of Tat-H10 fusion peptides could be 10-fold higher than Tat-H5 and Tat-H20 versions. Also, 10 His residues distributed at both sides of Tat peptide (C-5H-Tat-5H-C), are 102-fold more efficient than its equivalent C-Tat-10H-C. Although the optimal number of His residues is believed to be ranging between 5 and 10 (similar to the figures seen in His-based affinity tags, Table 1), the incorporation of only one His residue is sufficient to grant membrane-active properties to a drug delivery system [29]. Interestingly, although most of His-tags seem to favour intracellular targeting exclusively through proton sponge activities, LAH4 (KKALLALALHHLAHLALHLALALKKA) is believed to interact with the endosomal membrane at pH 7, due to its amphipathic nature, and disrupt its physical integrity upon subsequent protonation [30].

On the other hand, H6 has been recently found involved in intermolecular protein-protein interactions, leading to the self-assembling of C-terminal His-tagged building blocks into nanoparticles, useful as vehicles in non viral gene therapy [31, 32]. The nature of such organizing interactions is yet to be completely understood, and might involve other specific amino acids such arginines. Since the resulting nanoparticles seem to be more efficient in transgene expression when formed at slightly acidic pH values [32], its seems reasonable to speculate that protonated, overhanging H6 tails interact with anionic protein areas of siding building bocks and also with DNA. A deeper understanding of the interactions promoted by His-rich peptides could offer tools for the rational engineering of nanoparticle formation and DNA encapsulation, both issues of critical relevance in bionanomedicine [33, 34].

Conclusions

Protein production scientists are usually unaware of the potent biological properties of histidine-rich peptides for which they are used in nanomedicine (essentially endosomal disruption but also cross-molecular electrostatic interactions), which are activated at acidic pH. The same (or very similar) His-rich peptides used in affinity chromatography, incorporated into different type of nanoparticles (most ranging from H2 to H10, Table 2) to favour intracellular trafficking in drug delivery [25], are equally employed as affinity tags for separation and often not removed after protein purification (Table 1). These remaining agents might render unexpected protein behaviour, when exposed to cells or nucleic acids, and specific measures for control should be implemented in biological assays when technical obstacles make His-tag removal inconvenient upon protein purification.

References

  1. Waugh DS: Making the most of affinity tags. Trends Biotechnol. 2005, 23: 316-320. 10.1016/j.tibtech.2005.03.012

    Article  CAS  Google Scholar 

  2. Kimple ME, Sondek J: Overview of affinity tags for protein purification. Curr Protoc Protein Sci. 2004, Chapter 9: Unit 9.9,

    Google Scholar 

  3. Arnau J, Lauritzen C, Petersen GE, Pedersen J: Current strategies for the use of affinity tags and tag removal for the purification of recombinant proteins. Protein Expr Purif. 2006, 48: 1-13. 10.1016/j.pep.2005.12.002

    Article  CAS  Google Scholar 

  4. Waugh DS: An overview of enzymatic reagents for the removal of affinity tags. Protein Expr Purif. 2011, 80: 283-293. 10.1016/j.pep.2011.08.005

    Article  CAS  Google Scholar 

  5. Li Y: Self-cleaving fusion tags for recombinant protein production. Biotechnol Lett. 2011, 33: 869-881. 10.1007/s10529-011-0533-8

    Article  CAS  Google Scholar 

  6. Fong BA, Wood DW: Expression and purification of ELP-intein-tagged target proteins in high cell density E. coli fermentation. Microb Cell Fact. 2010, 9: 77- 10.1186/1475-2859-9-77

    Article  Google Scholar 

  7. Wood DW, Wu W, Belfort G, Derbyshire V, Belfort M: A genetic system yields self-cleaving inteins for bioseparations. Nat Biotechnol. 1999, 17: 889-892. 10.1038/12879

    Article  CAS  Google Scholar 

  8. Wu WY, Mee C, Califano F, Banki R, Wood DW: Recombinant protein purification by self-cleaving aggregation tag. Nat Protoc. 2006, 1: 2257-2262. 10.1038/nprot.2006.314

    Article  CAS  Google Scholar 

  9. Carson M, Johnson DH, McDonald H, Brouillette C, Delucas LJ: His-tag impact on structure. Acta Crystallogr D Biol Crystallogr. 2007, 63: 295-301. 10.1107/S0907444906052024

    Article  CAS  Google Scholar 

  10. Wang Z, Li H, Guan W, Ling H, Wang Z, Mu T, et al: Human SUMO fusion systems enhance protein expression and solubility. Protein Expr Purif. 2010, 73: 203-208. 10.1016/j.pep.2010.05.001

    Article  CAS  Google Scholar 

  11. Sun QM, Chen LL, Cao L, Fang L, Chen C, Hua ZC: An improved strategy for high-level production of human vasostatin120-180. Biotechnol Prog. 2005, 21: 1048-1052.

    Article  CAS  Google Scholar 

  12. Tang W, Sun ZY, Pannell R, Gurewich V, Liu JN: An efficient system for production of recombinant urokinase-type plasminogen activator. Protein Expr Purif. 1997, 11: 279-283. 10.1006/prep.1997.0800

    Article  CAS  Google Scholar 

  13. Nallamsetty S, Waugh DS: Solubility-enhancing proteins MBP and NusA play a passive role in the folding of their fusion partners. Protein Expr Purif. 2006, 45: 175-182. 10.1016/j.pep.2005.06.012

    Article  CAS  Google Scholar 

  14. Stefan A, Conti M, Rubboli D, Ravagli L, Presta E, Hochkoeppler A: Overexpression and purification of the recombinant diphtheria toxin variant CRM197 in Escherichia coli. J Biotechnol. 2011, 245-252. 156,

  15. Li Y: The tandem affinity purification technology: an overview. Biotechnol Lett. 2011, 33: 1487-1499. 10.1007/s10529-011-0592-x

    Article  CAS  Google Scholar 

  16. Li Y: Commonly used tag combinations for tandem affinity purification. Biotechnol Appl Biochem. 2010, 55: 73-83. 10.1042/BA20090273

    Article  CAS  Google Scholar 

  17. Hochuli E, Bannwarth W, Dobeli H, Gentz R, Stuber D: Genetic Approach to Facilitate Purification of Recombinant Proteins with a Novel Metal Chelate Adsorbent. Nat Biotech. 1988, 6: 1321-1325. 10.1038/nbt1188-1321. 10.1038/nbt1188-1321

    Article  CAS  Google Scholar 

  18. Block H, Kubicek J, Labahn J, Roth U, Schafer F: Production and comprehensive quality control of recombinant human Interleukin-1beta: a case study for a process development strategy. Protein Expr Purif. 2008, 57: 244-254. 10.1016/j.pep.2007.09.019

    Article  CAS  Google Scholar 

  19. Porath J, Carlsson J, Olsson I, Belfrage G: Metal chelate affinity chromatography, a new approach to protein fractionation. Nature. 1975, 258: 598-599. 10.1038/258598a0

    Article  CAS  Google Scholar 

  20. Porath J: Immobilized metal ion affinity chromatography. Protein Expr Purif. 1992, 3: 263-281. 10.1016/1046-5928(92)90001-D

    Article  CAS  Google Scholar 

  21. Gaberc-Porekar V, Menart V: Perspectives of immobilized-metal affinity chromatography. J Biochem Biophys Methods. 2001, 49: 335-360. 10.1016/S0165-022X(01)00207-X

    Article  CAS  Google Scholar 

  22. Hochuli E, Dobeli H, Schacher A: New metal chelate adsorbent selective for proteins and peptides containing neighbouring histidine residues. J Chromatogr. 1987, 411: 177-184.

    Article  CAS  Google Scholar 

  23. Knecht S, Ricklin D, Eberle AN, Ernst B: Oligohis-tags: mechanisms of binding to Ni2+-NTA surfaces. J Mol Recognit. 2009, 22: 270-279. 10.1002/jmr.941

    Article  CAS  Google Scholar 

  24. Patwardhan AV: Selection of optimum affinity tags from a phage-displayed peptide library Application to immobilized copper(II) affinity chromatography. J Chromatograph A. 1997, 91-100. 787,

    CAS  Google Scholar 

  25. Vazquez E, Ferrer-Miralles N, Villaverde A: Peptide-assisted traffic engineering for nonviral gene therapy. Drug Discov Today. 2008, 13: 1067-1074. 10.1016/j.drudis.2008.08.008

    Article  CAS  Google Scholar 

  26. Behr JP: The Proton Sponge: a Trick to Enter Cells the Viruses Did Not Exploit. Chimia. 1997, 51: 34-36.

    CAS  Google Scholar 

  27. Lo SL, Wang S: An endosomolytic Tat peptide produced by incorporation of histidine and cysteine residues as a nonviral vector for DNA transfection. Biomaterials. 2008, 29: 2408-2414. 10.1016/j.biomaterials.2008.01.031

    Article  CAS  Google Scholar 

  28. Midoux P, Kichler A, Boutin V, Maurizot JC, Monsigny M: Membrane permeabilization and efficient gene transfer by a peptide containing several histidines. Bioconjug Chem. 1998, 9: 260-267. 10.1021/bc9701611

    Article  CAS  Google Scholar 

  29. Kumar VV, Pichon C, Refregiers M, Guerin B, Midoux P, Chaudhuri A: Single histidine residue in head-group region is sufficient to impart remarkable gene transfection properties to cationic lipids: evidence for histidine-mediated membrane fusion at acidic pH. Gene Ther. 2003, 10: 1206-1215. 10.1038/sj.gt.3301979

    Article  CAS  Google Scholar 

  30. Kichler A, Leborgne C, Danos O, Bechinger B: Characterization of the gene transfer process mediated by histidine-rich peptides. J Mol Med. 2007, 85: 191-201. 10.1007/s00109-006-0119-4

    Article  CAS  Google Scholar 

  31. Vazquez E, Cubarsi R, Unzueta U, Roldan M, Domingo-Espin J, Ferrer-Miralles N, et al: Internalization and kinetics of nuclear migration of protein-only, arginine-rich nanoparticles. Biomaterials. 2010, 31: 9333-9339. 10.1016/j.biomaterials.2010.08.065

    Article  CAS  Google Scholar 

  32. Vazquez E, Roldan M, ez-Gil C, Unzueta U, Domingo-Espin J, Cedano J, et al: Protein nanodisk assembling and intracellular trafficking powered by an arginine-rich (R9) peptide. Nanomedicine (Lond). 2010, 5: 259-268. 10.2217/nnm.09.98. 10.2217/nnm.09.98

    Article  CAS  Google Scholar 

  33. Vazquez E, Villaverde A: Engineering building blocks for self-assembling protein nanoparticles. Microb Cell Fact. 2010, 9: 101- 10.1186/1475-2859-9-101

    Article  CAS  Google Scholar 

  34. Villaverde A: Nanotechnology, bionanotechnology and microbial cell factories. Microb Cell Fact. 2010, 9: 53- 10.1186/1475-2859-9-53

    Article  Google Scholar 

  35. Khan F, He M, Taussig MJ: Double-hexahistidine tag with high-affinity binding for protein immobilization, purification, and detection on ni-nitrilotriacetic acid surfaces. Anal Chem. 2006, 78: 3072-3079. 10.1021/ac060184l

    Article  CAS  Google Scholar 

  36. Jung HJ, Kim SK, Min WK, Lee SS, Park K, Park YC: Polycationic amino acid tags enhance soluble expression of Candida antarctica lipase B in recombinant Escherichia coli. Bioprocess Biosyst Eng. 2011, 833-839. 34,

    Article  CAS  Google Scholar 

  37. Hofstrom C, Orlova A, Altai M, Wangsell F, Graslund T, Tolmachev V: Use of a HEHEHE purification tag instead of a hexahistidine tag improves biodistribution of affibody molecules site-specifically labeled with (99m)Tc, (111)In, and (125)I. J Med Chem. 2011, 54: 3817-3826. 10.1021/jm200065e

    Article  CAS  Google Scholar 

  38. Schaefer G, Haber L, Crocker LM, Shia S, Shao L, Dowbenko D, et al: A Two-in-One Antibody against HER3 and EGFR Has Superior Inhibitory Activity Compared with Monospecific Antibodies. Cancer Cell. 2011, 20: 472-486. 10.1016/j.ccr.2011.09.003

    Article  CAS  Google Scholar 

  39. Cheng Z, Biechele T, Wei Z, Morrone S, Moon RT, Wang L, et al: Crystal structures of the extracellular domain of LRP6 and its complex with DKK1. Nat Struct Mol Biol. 2011, 18: 1204-1210. 10.1038/nsmb.2139

    Article  CAS  Google Scholar 

  40. Abramczyk O, Tavares CD, Devkota AK, Ryazanov AG, Turk BE, Riggs AF, et al: Purification and characterization of tagless recombinant human elongation factor 2 kinase (eEF-2K) expressed in Escherichia coli. Protein Expr Purif. 2011, 79: 237-244. 10.1016/j.pep.2011.05.005

    Article  CAS  Google Scholar 

  41. Yeliseev A, Zoubak L, Gawrisch K: Use of dual affinity tags for expression and purification of functional peripheral cannabinoid receptor. Protein Expr Purif. 2007, 53: 153-163. 10.1016/j.pep.2006.12.003

    Article  CAS  Google Scholar 

  42. Gehrmann M, Doss BT, Wagner M, Zettlitz KA, Kontermann RE, Foulds G, et al: A novel expression and purification system for the production of enzymatic and biologically active human granzyme B. J Immunol Methods. 2011, 371: 8-17. 10.1016/j.jim.2011.06.007

    Article  CAS  Google Scholar 

  43. Das KM, Banerjee S, Shekhar N, Damodaran K, Nair R, Somani S, et al: Cloning, Soluble Expression and Purification of High Yield Recombinant hGMCSF in Escherichia coli. Int J Mol Sci. 2011, 12: 2064-2076. 10.3390/ijms12032064

    Article  CAS  Google Scholar 

  44. McKenzie DL, Smiley E, Kwok KY, Rice KG: Low molecular weight disulfide cross-linking peptides as nonviral gene delivery carriers. Bioconjug Chem. 2000, 11: 901-909. 10.1021/bc000056i

    Article  CAS  Google Scholar 

  45. Midoux P, Kichler A, Boutin V, Maurizot JC, Monsigny M: Membrane permeabilization and efficient gene transfer by a peptide containing several histidines. Bioconjug Chem. 1998, 9: 260-267. 10.1021/bc9701611

    Article  CAS  Google Scholar 

  46. Ashley CE, Carnes EC, Phillips GK, Durfee PN, Buley MD, Lino CA, et al: Cell-specific delivery of diverse cargos by bacteriophage MS2 virus-like particles. ACS Nano. 2011, 5: 5729-5745. 10.1021/nn201397z

    Article  CAS  Google Scholar 

  47. Moore NM, Sheppard CL, Barbour TR, Sakiyama-Elbert SE: The effect of endosomal escape peptides on in vitro gene delivery of polyethylene glycol-based vehicles. J Gene Med. 2008, 10: 1134-1149. 10.1002/jgm.1234

    Article  CAS  Google Scholar 

  48. Tarwadi , Jazayeri JA, Prankerd RJ, Pouton CW: Preparation and in vitro evaluation of novel lipopeptide transfection agents for efficient gene delivery. Bioconjug Chem. 2008, 19: 940-950. 10.1021/bc700463q

    Article  CAS  Google Scholar 

  49. Kichler A, Leborgne C, Danos O, Bechinger B: Characterization of the gene transfer process mediated by histidine-rich peptides. J Mol Med. 2007, 85: 191-201. 10.1007/s00109-006-0119-4

    Article  CAS  Google Scholar 

  50. Chang KL, Higuchi Y, Kawakami S, Yamashita F, Hashida M: Efficient gene transfection by histidine-modified chitosan through enhancement of endosomal escape. Bioconjug Chem. 2010, 21: 1087-1095. 10.1021/bc1000609

    Article  CAS  Google Scholar 

  51. Chang KL, Higuchi Y, Kawakami S, Yamashita F, Hashida M: Development of lysine-histidine dendron modified chitosan for improving transfection efficiency in HEK293 cells. J Control Release. 2011, 156: 195-202. 10.1016/j.jconrel.2011.07.021

    Article  CAS  Google Scholar 

  52. Yamano S, Dai J, Yuvienco C, Khapli S, Moursi AM, Montclare JK: Modified Tat peptide with cationic lipids enhances gene transfection efficiency via temperature-dependent and caveolae-mediated endocytosis. J Control Release. 2011, 152: 278-285. 10.1016/j.jconrel.2011.02.004

    Article  CAS  Google Scholar 

  53. Gu J, Wang X, Jiang X, Chen Y, Chen L, Fang X, et al: Self-assembled carboxymethyl poly (l-histidine) coated poly (beta-amino ester)/DNA complexes for gene transfection. Biomaterials. 2012, 33: 644-658. 10.1016/j.biomaterials.2011.09.076

    Article  CAS  Google Scholar 

  54. Tanaka K, Kanazawa T, Ogawa T, Suda Y, Takashima Y, Fukuda T, et al: A novel, bio-reducible gene vector containing arginine and histidine enhances gene transfection and expression of plasmid DNA. Chem Pharm Bull (Tokyo). 2011, 59: 202-207. 10.1248/cpb.59.202. 10.1248/cpb.59.202

    Article  CAS  Google Scholar 

  55. Hatefi A, Megeed Z, Ghandehari H: Recombinant polymer-protein fusion: a promising approach towards efficient and targeted gene delivery. J Gene Med. 2006, 8: 468-476. 10.1002/jgm.872

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We appreciate the financial support received for the design and microbial production of artificial viruses from MICINN (ACI2009-0919), FISS (PS09/00165), AGAUR (2009SGR-108) and CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), an initiative funded by the VI National R&D&i Plan 2008-2011, Iniciativa Ingenio 2010, Consolider Program, CIBER Actions and financed by the Instituto de Salud Carlos III with assistance from the European Regional Development Fund. KCG and PK gratefully acknowledge the financial support from Indo-Spanish Joint Project. AV has been distinguished with an ICREA ACADEMIA award.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Antonio Villaverde.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

NFM and JLC have equally contributed to this manuscript. All authors have read and approved the final version.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Ferrer-Miralles, N., Corchero, J.L., Kumar, P. et al. Biological activities of histidine-rich peptides; merging biotechnology and nanomedicine. Microb Cell Fact 10, 101 (2011). https://doi.org/10.1186/1475-2859-10-101

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1475-2859-10-101

Keywords