Acessibilidade / Reportar erro

Molecular farming of antimicrobial peptides: available platforms and strategies for improving protein biosynthesis using modified virus vectors

Abstract

The constant demand for new antibiotic drugs has driven efforts by the scientific community to prospect for peptides with a broad spectrum of action. In this context, antimicrobial peptides (AMPs) have acquired great scientific importance in recent years due to their ability to possess antimicrobial and immunomodulatory activity. In the last two decades, plants have attracted the interest of the scientific community and industry as regards their potential as biofactories of heterologous proteins. One of the most promising approaches is the use of viral vectors to maximize the transient expression of drugs in the leaves of the plant Nicotiana benthamiana. Recently, the MagnifectionTM expression system was launched. This sophisticated commercial platform allows the assembly of the viral particle in leaf cells and the systemic spread of heterologous protein biosynthesis in green tissues caused by Agrobacterium tumefaciens “gene delivery method”. The system also presents increased gene expression levels mediated by potent viral expression machinery. These characteristics allow the mass recovery of heterologous proteins in the leaves of N. benthamiana in 8 to 10 days. This system was highly efficient for the synthesis of different classes of pharmacological proteins and contains enormous potential for the rapid and abundant biosynthesis of AMPs.

Key words
antimicrobial peptides; Magnifection; Nicotiana benthamiana; transient expression

INTRODUCTION

Historically, pathogenic microorganisms represent the greatest health risk for individuals whose immune system is compromised by milder diseases or by invasive therapies (Watkins and Bonomo 2016WATKINS RR and BONOMO RA. 2016. Overview: Global and Local Impact of Antibiotic Resistance. Infect Dis Clin North Am 30: 313-322.). The need to attenuate or even prevent the proliferation of these microorganisms in the hospital environment has been a constant concern of leading scientific research groups around the world (Uhlig et al. 2014UHLIG T, KYPRIANOU T, MARTINELLI FG, OPPICI CA, HEILIGERS D, HILLS D, CALVO XR and VERHAERT P. 2014. The emergence of peptides in the pharmaceutical business: From exploration to exploitation. EuPA Open Proteomics 4: 58-69.). Many of the pathogens commonly found in hospitals are resistant to antimicrobial drugs commonly used in the treatment of common or high-risk conditions. The indiscriminate use of antibiotics and antifungals without a medical prescription has led, since the mid-1960s, to a consistent increase in the isolation of resistant strains in hospitals, especially in Latin America (Andersson et al. 2016ANDERSSON DI, HUGHES D and KUBICEK-SUTHERLAND JZ. 2016. Mechanisms and consequences of bacterial resistance to antimicrobial peptides. Drug Resist Updat 26: 43-57.).

The high rate with which resistant pathogens are selected throughout the treatment of patients is still the main problem regarding the use of conventional antimicrobials. In this scenario, many of the most common, cheap and once-effective pathogen control drugs become ineffective in systemic infection (Perry et al. 2016). Due to the exponential growth of cases of antibiotic-resistant microorganisms, the development of new control alternatives and molecules with broad action spectrum and low toxicity has acquired a strategic character for the pharmaceutical industry (Kosikowska and Lesner 2016KOSIKOWSKA P and LESNER A. 2016. Antimicrobial peptides (AMPs) as drug candidates: a patent review (2003–2015). Expert Opin Ther Pat 26: 689-702., Nagel et al. 2016NAGEL TE, CHAN BK, DE VOS D, EL-SHIBINY A, KANG’ETHE EK, MAKUMI A and PIRNAY J-P. 2016. The Developing World Urgently Needs Phages to Combat Pathogenic Bacteria. Front Microbiol 7: 882., Andersson et al. 2016ANDERSSON DI, HUGHES D and KUBICEK-SUTHERLAND JZ. 2016. Mechanisms and consequences of bacterial resistance to antimicrobial peptides. Drug Resist Updat 26: 43-57.).

In this context, antimicrobial peptides (AMPs) present themselves as a valuable alternative in the complementation or substitution of traditional therapeutic compounds. AMPS are natural antibiotics found in microorganisms, plants and animals, which function as important components of the innate immune system, the first line of defense of these organisms against exogenous pathogens. In the early stages of infection, molecular stimuli emitted by intrinsic signaling systems trigger the biosynthesis of AMPS in order to cope with the invading agent (Mojsoska and Jenssen 2015MOJSOSKA B and JENSSEN H. 2015. Peptides and Peptidomimetics for Antimicrobial Drug Design. Pharmaceuticals 8: 366-415.).

From the structural point of view, AMPs are characterized as small protein molecules with less than 100 amino acid residues, often rich in cysteine, glycine or histidine residues and which can be classified according to the presence of α-helices, β-sheets, extensions in the side chains of the nucleus amino acids or by the formation of disordered loops (Cunha et al. 2017CUNHA NB et al. 2017. The next generation of antimicrobial peptides (AMPs) as molecular therapeutic tools for the treatment of diseases with social and economic impacts. Drug Discov Today 22: 234-248.). These small peptides can also be grouped according to their physicochemical properties in anionic, cationic (usually with positive charges between 2 to 9) and amphipathic AMPs, with their hydrophobic and hydrophilic portions placed side by side (Nguyen et al. 2011NGUYEN LT, HANEY EF and VOGEL HJ. 2011. The expanding scope of antimicrobial peptide structures and their modes of action. Trends Biotechnol 29: 464-472., Malanovic and Lohner 2016MALANOVIC N and LOHNER K. 2016. Gram-positive bacterial cell envelopes: The impact on the activity of antimicrobial peptides. Biochim Biophys Acta 1858: 936-946.).

The first function of AMPs is to kill or inhibit the growth of microorganisms at the molecular level. In general, the classical mode of action of cationic AMPs is due to the structural disorganization and formation of pores in the membrane, with the consequent rupture of the lipid bilayer that delimits fungal cells and Gram positive and negative bacteria (Perry et al. 2016). For this reason, AMPs can act against different cell targets and in some cases, be considered “promiscuous” molecules, capable of performing more than one function, regardless of their structure. Recent reports have shown that some AMPs can destabilize translational machinery (ribosomes) and inhibit DNA synthesis and the cell cycle of certain fungi and bacteria (Nguyen et al. 2011, Malanovic and Lohner 2016MALANOVIC N and LOHNER K. 2016. Gram-positive bacterial cell envelopes: The impact on the activity of antimicrobial peptides. Biochim Biophys Acta 1858: 936-946.). Some of these molecules can still act as antivirals (Malanovic and Lohner 2016), as insecticides and as hemolytic and antitumor agents (Nguyen et al. 2011NGUYEN LT, HANEY EF and VOGEL HJ. 2011. The expanding scope of antimicrobial peptide structures and their modes of action. Trends Biotechnol 29: 464-472., 2012NGUYEN GKT, LIM WH, NGUYEN PQT and TAM JP. 2012. Novel Cyclotides and Uncyclotides with Highly Shortened Precursors from Chassalia chartacea and Effects of Methionine Oxidation on Bioactivities. J Biol Chem 287: 17598-17607.).

The biochemical and physicochemical characteristics inherent to the structure of the AMPs are crucial for the types of biological functions of these molecules. The length and distribution of charged groups along the peptide chain, degree of hydrophobicity, N-terminal amidation, and the type and number of secondary structures determine the efficiency of the bioactive peptide and its cytotoxic ability to different cell types and microorganisms (Nguyen et al. 2011).

Among the several AMPs already characterized, one class in particular has a special value for medicine: the innate defense peptides (host defense peptides) (HDPs). These small 12- to 50-amino acid peptides are generally non-toxic to mammalian cells and can be isolated from unicellular, invertebrate microorganisms such as mollusks, plants, amphibians, birds, fish and mammals (including humans). HDPs conjugate the direct antimicrobial action with the modulation of the innate immunity of the host organism through the induction of the inflammatory process. Thus, when synthesized in response to microbial action, HDPs activate cytokine synthesis, the occurrence of apoptosis and chemotaxis, the recruitment and proliferation of macrophages, neutrophils, eosinophils and T lymphocytes, as well as differentiation of dendritic cells (Silva et al. 2011SILVA ON, MULDER KC, BARBOSA AE, OTERO-GONZALEZ AJ, LOPEZ-ABARRATEGUI C and REZENDE TM. 2011. Exploring the pharmacological potential of promiscuous host-defense peptides: from natural screenings to biotechnological applications. Front Microbiol 22: 2;232.).

The diverse functions that the HDPs can simultaneously present and their degree of selectivity reflect the enormous potential of use of these molecules for the protection of patients. This phenomenon of “promiscuity and functional dynamism” is characterized as the basic principle for the design of new drugs and for the preparation of biotechnological studies with AMPs candidates to act as new antibiotics (Pelegrini et al. 2007PELEGRINI PB, QUIRINO BF and FRANCO OL. 2007. Plant cyclotides: An unusual class of defense compounds. Peptides 28: 1475-1481.).

THE HETEROLOGOUS EXPRESSION OF AMPS AIMED AT IMPROVING THE FIGHT AGAINST FUNGAL AND BACTERIAL INFECTIONS

The study and the pharmaceutical evaluation of proteins (among them the AMPs), in pre-commercial character, requires the obtaining of these molecules in great quantities. For the assessment of aspects such as the different mechanisms of action and the structure/function relation, masses of peptides ranging from a few micrograms to many milligrams are required. In this context, the large-scale production of AMPs is the greatest challenge in prospecting and characterization studies of these peptides (Parachin et al. 2012PARACHIN NS, MULDER KC, VIANA AA, DIAS SC and FRANCO OL. 2012. Expression systems for heterologous production of antimicrobial peptides. Peptides:38(2): 446-456.).

The method of obtaining AMPs considered more advantageous in terms of cost and yield is one that explores recombinant or biofactory systems for the synthesis of heterologous proteins. In the last decades, several systems of heterologous expression have been developed with peculiarities that allow high levels of synthesis, modification and secretion of heterologous proteins (Parachin et al. 2012PARACHIN NS, MULDER KC, VIANA AA, DIAS SC and FRANCO OL. 2012. Expression systems for heterologous production of antimicrobial peptides. Peptides:38(2): 446-456., Parachin and Franco 2014PARACHIN NS and FRANCO OL. 2014. New edge of antibiotic development: antimicrobial peptides and corresponding resistance. Front Microbiol 5: 147.). This approach also allows the structural modification of peptides to carry out detailed studies on the function of these molecules.

More than 95% of the heterologous peptides are synthesized in heterologous expression systems using bacterial or yeast cells (Parachin et al. 2012). In these systems, the bacterial species most commonly used is Escherichia coli, a popular host because of its rapid growth, compatibility with a wide variety of expression vectors, low cost and high control of gene expression due to the wide knowledge about its genetic mechanisms and its physiology (Parachin and Franco 2014).

The isolation and prospection of several AMPs have been largely carried out by the group of researchers from the Center for Proteomic and Biochemical Analysis of the Catholic University of Brasilia (CAPB UCB). In addition to the structural and functional characterization of AMPs, the CAPB group specialized in its heterologous expression in bacterial system and in yeast cells. CAPB researchers have been successful in isolating the peptides from their natural sources or by their in silico redesign, which allows the addition, removal or replacement of amino acids with the aim of increasing antimicrobial activity or adding characteristics such as immunomodulatory capacity.

PROMISING AMPS WITH POTENTIAL THERAPEUTIC PROPERTIES AGAINST MULTIDRUG RESISTANT PATHOGENS

The antimicrobial peptide Cn-AMP1 was first isolated from coconut water (Cocos nucifera) in 2009, by Mandal and colleagues (Mandal et al. 2009MANDAL SM, DEY S, MANDAL M, SARKAR S, MARIA-NETO S and FRANCO OL. 2009. Identification and structural insights of three novel antimicrobial peptides isolated from green coconut water. Peptides 30: 633-637.). This small peptide is part of a peptide family with two other AMPs, called Cn-AMPs 2 and 3. All present nine amino acid residues with molecular masses between 0.8 and 1.3 kDa. When compared to the other two, Cn-AMP1 is the most active in vitro against the bacteria Klebsiella pneumoniae and Pseudomonas aeruginosa (MIC: 9 μM) and the fungi Candida albicans, Trichophyton rubrum, an anthropophilic dermatophyte fungus very common in skin diseases such as athlete’s foot, and Trichophyton mentagrophytes (MIC: 18 μM) (Mandal et al. 2009MANDAL SM, DEY S, MANDAL M, SARKAR S, MARIA-NETO S and FRANCO OL. 2009. Identification and structural insights of three novel antimicrobial peptides isolated from green coconut water. Peptides 30: 633-637., Santana et al. 2015SANTANA MJ, DE OLIVEIRA AL, QUEIROZ JÚNIOR LHK, MANDAL SM, MATOS CO, DE O. DIAS R, FRANCO OL and LIÃO LM. 2015. Structural insights into Cn-AMP1, a short disulfide-free multifunctional peptide from green coconut water. FEBS Lett 589: 639-644.). Other in vitro biological activity assays have shown that Cn-AMP1 exerts several alternative functions simultaneously to microbial control. The peptide was able to reduce the viability of different types of tumor cells, besides promoting immunomodulatory activity through the stimulation of cytokine secretion by laboratory-grown monocytes, without causing cytotoxicity to human erythrocytes and murine macrophages (Silva et al. 2012SILVA ON et al. 2012. Cn-AMP1: A new promiscuous peptide with potential for microbial infections treatment. Pept Sci 98: 322-331.).

Another potent interesting antimicrobial peptide is Cm-p5, isolated from the mollusk Centrichis muricatus. Its prospection was performed after the tryptic fragmentation of a chromatographic fraction containing isolated peptides of C. muricatus analyzed by MS/MS, which generated a new candidate sequence (López-Abarrategui et al. 2015LÓPEZ-ABARRATEGUI C et al. 2015. Cm-p5: an antifungal hydrophilic peptide derived from the coastal mollusk Cenchritis muricatus (Gastropoda: Littorinidae). FASEB J 29: 3315-3325.). From bioinformatics analyzes, a series of variant peptides was theoretically proposed based on the sequence, and these were previously evaluated against C. albicans fungus. Of these, Cm-p5 has demonstrated remarkable antifungal activity against strains of C. albicans with high medical importance. For purposes of illustration, the preliminary results of activity demonstrated MICs of 1 and 0.5 μg.mL-1, enough to obtain therapeutic effects. In addition, this peptide was also extremely active against Cryptococcus neoformans and T. rubrum. These values far exceed the activity of its precursor Cm-p1, reported in 2011. These results were the first to open the way for toxicity tests in animal cells and for the study of their mechanism of action (López-Abarrategui et al. 2015).

Clavanin A, a highly efficient AMP in the control of bacteria such as P. aeruginosa, was recently characterized by our group and demonstrated extremely high activity in the control of Gram positive and negative bacteria both in vitro and in vivo, from assays with mice (data in press). In addition, clavanin A was also efficient as a molecular stimulating agent for defense reactions of the attacked organisms (Silva et al. 2015SILVA ON, FENSTERSEIFER ICM, RODRIGUES EA, HOLANDA HHS, NOVAES NRF and CUNHA JPA. 2015. Clavanin A improves outcome of complications from different bacterial infections. Antimicrob Agents Chemother 59(3): 1620-1626.). Therefore, the therapeutic potential of Clavanin A is promising, both in the direct antimicrobial action and in the immunomodulatory activity without aggravating the inflammatory response.

Among the AMPs isolated from plants, cyclotides are one of the most promising biomolecules with therapeutic potential against different classes of plant pathogens. These peptides were first described in the 1970s, isolated from different tissues such as leaves, stems, flowers, roots and barks of various plant families, such as Violaceae, Rubiaceae, Fabaceae, Asteraceae and Cucurbitaceae (Chen et al. 2005CHEN GH, HSU MP, TAN CH, SUNG HY, KUO CG, FAN MJ, CHEN HM, CHEN S and CHEN CS. 2005. Cloning and Characterization of a Plant Defensin VaD1 from Azuki Bean. J Agric Food Chem 53: 982-988., Craik et al. 2006CRAIK DJ, ČEMAŽAR M, WANG CKL and DALY NL. 2006. The cyclotide family of circular miniproteins: Nature’s combinatorial peptide template. J Pept Sci 84: 250-266.). The cyclotides are rich in cysteine and have between 28 and 37 amino acid residues, and have a unique characteristic: a cyclic structure, where the N-terminal region is linked to the C-terminal region in a circular chain, which distinguishes them (CisI-CisIV, CisII-CisV, CisIII-CisVI), and the presence of a single, highly conserved three-disulphide pattern.

This arrangement forms a motif known as Cyclic Cysteine Knot (CCK), which comprises a peptidic ring formed by two parallel disulphide bridges (CisI-CisIV and CisII-CisV) and a transverse disulphide bridge (CisIII-CisVI), which stabilize the motif. In this way, from the combining of the cyclization and the CCK motif, the family of cyclotides groups vegetable cyclic peptides that share a three-dimensional structure that gives the molecules high stability and protection against proteolytic enzymes, as well as acidic conditions and chemical and physical degradation (Craik et al. 2006CRAIK DJ, ČEMAŽAR M, WANG CKL and DALY NL. 2006. The cyclotide family of circular miniproteins: Nature’s combinatorial peptide template. J Pept Sci 84: 250-266., Pelegrini et al. 2007PELEGRINI PB, QUIRINO BF and FRANCO OL. 2007. Plant cyclotides: An unusual class of defense compounds. Peptides 28: 1475-1481., Pinto et al. 2016PINTO MFS et al. 2016. Characterization of a Bioactive Acyclotide from Palicourea rigida. J Nat Prod 79: 2767-2773., Cunha et al. 2016CUNHA NBD et al. 2016. Cloning and characterization of novel cyclotides genes from South American plants. J Pept Sci 106: 784-795.).

Reports in the literature indicate the presence of large numbers of cyclotides in the Rubiaceae family and in a particular plant of this family, Palicourea condensata (Pinto et al. 2016). Cyclotides of a species of the central southern plateau of the same genus, Palicourea rigida, have been extensively studied by our group (Pinto et al. 2016). Curiously, this plant was little studied despite its widespread use in folk medicine in the region. Analysis of the protein content of P. rigida evidenced the presence of cyclotides in the inflorescence, peduncle and leaves, some of which appeared only in specific tissues, while others appeared in all explored parts. One of these peptides was sequenced and pioneered: Parigidin-br1, which contains 32 amino acid residues. This cyclotide was classified in the Bracelets subfamily due to the absence of a cis-proline in loop 5, and its function was evaluated in vivo against Lepidoptera (Diatraea saccharalis) neonates, and in vitro against insect SF-9 cells, presenting significant insecticidal and cytotoxic activities (Pinto et al. 2016). The scarcity of biochemical studies on P. rigida, added to its wide medicinal use, intensifies the interest for its evaluation as a source of Parigidina-br1, a molecule with clear potential of use for the protection of cultivated plants.

EXPRESSION OF RECOMBINANT DRUGS IN PLANTS - “MOLECULAR FARMING”

Since the development of recombinant DNA technology in the early 1970s, the recombinant production of proteins of pharmacological and industrial interest has been based on microbial (prokaryotic and eukaryotic) fermentation systems and culture of transgenic mammalian cells. Although these systems are highly efficient in terms of yield, they have some disadvantages in terms of authenticity (amino acid sequence identity and post-translational modifications), biosafety, and production costs, despite the recent development of strategies to minimize or cure them (Parachin et al. 2012PARACHIN NS, MULDER KC, VIANA AA, DIAS SC and FRANCO OL. 2012. Expression systems for heterologous production of antimicrobial peptides. Peptides:38(2): 446-456., da Cunha et al. 2014DA CUNHA NB, VIANNA GR, DA ALMEIDA LIMA T and RECH E. 2014. Molecular farming of human cytokines and blood products from plants: Challenges in biosynthesis and detection of plant-produced recombinant proteins. Biotechnol J 9: 39-50.).

One of the most promising alternatives for replacing the already established recombinant platforms is the use of plant systems as vehicles for the production of proteins used as diagnostic reagents, vaccines and drugs, also called plant-made pharmaceuticals (PMPs) or “drugs produced in Plants “ (Tregoning et al. 2005TREGONING JS, CLARE S, BOWE F, EDWARDS L, FAIRWEATHER N, QAZI O, NIXON PJ, MALIGA P, DOUGAN G and HUSSELL T. 2005. Protection against tetanus toxin using a plant-based vaccine. Eur J Immunol 35: 1320-1326., Cunha et al. 2017CUNHA NB et al. 2017. The next generation of antimicrobial peptides (AMPs) as molecular therapeutic tools for the treatment of diseases with social and economic impacts. Drug Discov Today 22: 234-248.). This application represents a relatively recent strand of biotechnology called Plant Molecular Farming, the scope of which is the synthesis of heterologous polypeptides of pharmaceutical interest using the plant reactor machinery (Ma et al. 2005MA JKC, CHIKWAMBA R, SPARROW P, FISCHER R, MAHONEY R and TWYMAN RM. 2005. Plant-derived pharmaceuticals - the road forward. Trends Plant Sci 10: 580-585.).

Plants can be attractive systems for the mass biosynthesis of drugs. Many plant species present desirable economic and qualitative aspects associated to the synthesis of “PMPs”, such as low production costs and capacity to perform post-translational modifications required by complex AMPs. Plant reactors or “plant biofactories” are potentially one of the most economical systems for the large-scale production of this class of biomolecules. The average cost of production of “PMPs” represents about 10% of the total presented by systems based on yeast cells, being able to be up to 50 times smaller than that of bacterial systems. When compared to sophisticated and relatively expensive platforms such as mammalian cell culture, this difference may be even greater. It is estimated that the cost of producing a given recombinant drug equivalently expressed in tobacco seeds and mammalian cell suspension may be up to 1000 times lower in the former than in the latter (Obembe et al. 2011).

The plants represent one of the types of biomass of lower cost of production in nature, requiring only soil, water and light for the production of large amounts of biomass, represented mainly by leaves and seeds. In contrast, cell culture of microorganisms and mammals requires the adoption of expensive fermenters and their manipulation under sterile conditions, something that considerably impairs the production process (Obembe et al. 2011OBEMBE OO, POPOOLA JO, LEELAVATHI S and REDDY SV. 2011. Advances in plant molecular farming. Biotechnol Adv 29: 210-222., Davies 2010DAVIES HM. 2010. Commercialization of whole-plant systems for biomanufacturing of protein products: evolution and prospects. Plant Biotechnol J 8: 845-861.).

Since 1986, when the first relevant recombinant drug has been synthesized in transgenic tobacco plants - human growth hormone (hGH) - more than one hundred different heterologous proteins, including antibodies, antigens, hormones, molecular transporters, anticoagulant peptides and enzymes synthesized in different plant systems (Cunha et al. 2011aCUNHA NB et al. 2011a. Expression of functional recombinant human growth hormone in transgenic soybean seeds. Transgenic Res 20: 811-826., bCUNHA NB et al. 2011b. Accumulation of functional recombinant human coagulation factor IX in transgenic soybean seeds. Transgenic Res 20: 841-855.). The commonly used platforms to produce all these classes of molecules are the suspension of tobacco cells; cereal and legume seeds, such as maize and soya; oilseed, such as canola; potato tubers; the different species of leafy crops, such as lettuce and spinach, as well as fruits such as tomatoes and bananas (Egelkrout et al. 2012EGELKROUT E, RAJAN V and HOWARD JA. 2012. Overproduction of recombinant proteins in plants. Plant Sci 184: 83-101., Ma et al. 2003MA JKC, DRAKE PMW and CHRISTOU P. 2003. The production of recombinant pharmaceutical proteins in plants. Nat Rev Genet 4: 794-805., Drake et al. 2017DRAKE PMW, SZETO TH, PAUL MJ, TEH AYH and MA JKC. 2017. Recombinant biologic products versus nutraceuticals from plants – a regulatory choice? Br J Clin Pharmacol 83: 82-87.).

In addition to economic factors, the plants present very promising aspects regarding the quality of heterologous proteins and the biosafety of their use as reactors of PMPs. The main technical aspects of the production of therapeutic proteins in the different available systems are compared in Table I.

TABLE I
Comparison between different production systems of heterologous proteins of pharmaceutical interest.

About thirty PMPs, including vaccines, antibodies and therapeutic proteins, have now reached the final stages of international pre-marketing protocols and tests. Eight recombinant plant-derived products - seven laboratory reagents and one drug - have already been released. In 2012, the recombinant enzyme glucocerebrosidase - or taliglucerase alfa, secreted by suspension carrot cells and used for the treatment of Gaucher’s neurodegenerative disease, became the first plant-biotech synthesized product to be released for commercialization in the United States (Tregoning et al. 2005TREGONING JS, CLARE S, BOWE F, EDWARDS L, FAIRWEATHER N, QAZI O, NIXON PJ, MALIGA P, DOUGAN G and HUSSELL T. 2005. Protection against tetanus toxin using a plant-based vaccine. Eur J Immunol 35: 1320-1326., Shaaltiel et al. 2015SHAALTIEL Y, GINGIS–VELITSKI S, TZABAN S, FIKS N, TEKOAH Y and AVIEZER D. 2015. Plant-based oral delivery of β-glucocerebrosidase as an enzyme replacement therapy for Gaucher’s disease. Plant Biotechnol J 13: 1033-1040., Drake et al. 2017DRAKE PMW, SZETO TH, PAUL MJ, TEH AYH and MA JKC. 2017. Recombinant biologic products versus nutraceuticals from plants – a regulatory choice? Br J Clin Pharmacol 83: 82-87.).

Basically, there are two ways of expressing genes to produce PMPs: i) the stable transformation of the nuclear or plastidial genomes and ii) the transient expression of genes delivered by Agrobacterium or infectious transgenic viruses (Obembe et al. 2011).

Stable transformation of the plant genome is the most commonly used strategy for the production of recombinant PMPs and is applicable to a wide variety of plant species: from cereals to legumes, including leafy crops, oleaginous and fruit plants. This method advocates the stable integration of transgenes into the genome of the host plant and allows the continuous synthesis of the recombinant protein as an inheritable phenotypic trait, either generically or localized in a specific plant organ (Xu et al. 2012XU J, DOLAN MC, MEDRANO G, CRAMER CL and WEATHERS PJ. 2012. Green factory: Plants as bioproduction platforms for recombinant proteins. Biotechnol Adv 30: 1171-1184.).

Transient gene expression is mediated by a bacterial or viral infective intermediate and is generally used to verify the efficiency of the gene construct activity employed in genetic transformation experiments and to quickly validate the structure and function of the recombinant protein (Loh et al. 2017LOH H-S, GREEN BJ and YUSIBOV V. 2017. Using transgenic plants and modified plant viruses for the development of treatments for human diseases. Curr Opin in Virol 26: 81-89.).

However, infiltration of plant leaves - notably the Nicotiana benthamiana species - by vacuum or syringes using recombinant Agrobacterium tumefaciens suspensions may result in the transient transformation of several leaf cells and the attainment of high levels of gene expression a few days after the realization of the experiment, providing the routinely scheduling of PMP production at industrial levels (Gleba et al. 2014GLEBA YY, TUSÉ D and GIRITCH A 2014. Plant Viral Vectors for Delivery by Agrobacterium. In: Palme, K and Gleba Y (Eds), Plant Viral Vectors, Berlin, Heidelberg: Springer Berlin Heidelberg, p. 155-192.). Table II shows the main comparative characteristics between the two strategies of genetic transformation of plants for the development of PMPs, as well as the platforms available in each expression scenario:

TABLE II
Available platforms for the biosynthesis of PMPs.

The selection of the plant species destined to the production of PMPs represents one of the most important criteria aiming at the success of the molecular strategy of gene expression. Genomic, biochemical, physiological and even morphological features inherent in each species have a crucial influence on a number of factors, such as the yield of recombinant proteins, the ability to promote post-translational modifications in complex proteins, the structural stability of the polypeptide and the final cost of production (Stoger at al. 2002STOGER E, SACK M, PERRIN Y, VAQUERO C, TORRES E, TWYMAN RM, CHRISTOU P and FISCHER R. 2002. Practical considerations for pharmaceutical antibody production in different crop systems. Molecular Breeding 9: 149-158., Fischer et al. 2004FISCHER R, STOGER E, SCHILLBERG S, CHRISTOU P and TWYMAN RM. 2004. Plant-based production of biopharmaceuticals. Curr Opin Plant Biol 7: 152-158.).

Table III illustrates the properties and peculiarities of the main plant species used as recombinant protein bioreactors for the pharmaceutical and industrial sectors.

TABLE III
Plant species used as bioreactors of PMPs.

STRATEGIES FOR MAXIMIZING RECOMBINANT PROTEIN YIELD IN PLANTS

One of the most important factors determining the viability of producing heterologous proteins in plants is to obtain satisfactory amounts of the polypeptides. The absolute yield of production depends on the maximization of efficiency of all stages of gene expression and protein stability. Thus, the various strategies aimed at increasing the final amount of recombinant protein in plant reactors focus on gene transcription, post-transcriptional processing, translation and post-translational protein stability (Streatfield 2007STREATFIELD SJ. 2007. Approaches to achieve high-level heterologous protein production in plants. Plant Biotechnol J 5: 2-15.).

STRATEGIES AT THE TRANSCRIPTIONAL LEVEL

Choice of promoters and terminators

To obtain high levels of transgene expression the two most important elements are the promoter and the transcription terminator (Ma et al. 2003MA JKC, DRAKE PMW and CHRISTOU P. 2003. The production of recombinant pharmaceutical proteins in plants. Nat Rev Genet 4: 794-805.).

Strong eukaryotic promoters present specific sites that facilitate the recognition of transcription factors and the RNA polymerase II. They can also be upregulated by specific enhancers that boost heterologous expression, providing high levels of mRNA production (Streatfield 2007STREATFIELD SJ. 2007. Approaches to achieve high-level heterologous protein production in plants. Plant Biotechnol J 5: 2-15., Tschofen et al. 2016TSCHOFEN M, KNOPP D, HOOD E and STÖGER E. 2016. Plant Molecular Farming: Much More than Medicines. Annu Rev Analyt Chem 9: 271-294.).

The strongest constitutive promoters most widely used for heterologous expression in dicotyledons are 19S and 35S CaMV, derived from the 19S and 35S transcripts of cauliflower mosaic virus (CaMV).

In monocots, the ubiquitin 1 (ubi-1) promoter from maize is also very popular and efficient for the expression of recombinant proteins in cereals (Twyman et al. 2003TWYMAN RM, STOGER E, SCHILLBERG S, CHRISTOU P and FISCHER R. 2003. Molecular farming in plants: host systems and expression technology. Trends Biotechnol 21: 570-578.).

Also, inducible promoters that allow external regulation by chemical and physical stimuli emerge as interesting tools to maximize gene expression at the transcriptional level (Twyman et al. 2003).

In this context, a peroxidase-activated sweet potato (Ipomoea batatas) promoter was used to enhance 30 times the expression of the gus gene in transgenic tobacco plants subjected to the presence of hydrogen peroxide and ultraviolet light than the 35S CaMV promoter (Kim et al. 2003KIM KY, KWON SY, LEE HS, HUR Y, BANG JW and KWAK SS. 2003. A novel oxidative stress-inducible peroxidase promoter from sweetpotato: molecular cloning and characterization in transgenic tobacco plants and cultured cells. Plant Mol Biol 51: 831-838.). Another interesting example was the rapid induction of biosynthesis of heterologous proteins in tobacco using the endogenous promoter that controls the gene of the enzyme hydroxy-3-methylglutaryl-Coa-reductase (HMGR2), activated by mechanical stress induced by harvesting practices, a system developed by the late american company Crop Tech Corp (Padidam 2003PADIDAM M. 2003. Chemically regulated gene expression in plants. Curr Opin Plant Biol 6: 169-177.).

Terminators widely used include the 35S CaMV, that of the genes nos and ssu, respectively of A. tumefaciens and pea (Pisum sativum) (Ma et al. 2003).

Minimization of transcriptional silencing

Several mechanisms lead to the interruption or inhibition of mRNA synthesis in plants, such as the presence of prokaryotic DNA sequences, typical of the plasmid backbone used in genetic transformation, DNA methylation, site-related “position effect” transgene integration into the plant genome, integration site structure, presence of multiple copies or “superfluous” copies of the transgene, sequences with potential for hairpin formation and double-stranded RNA, in addition to negative feedback from promoters - a common event when end products of gene expression are recombinant enzymes (Finnegan and McElroy 1994FINNEGAN J and MCELROY D. 1994. Transgene Inactivation: Plants Fight Back! Bio/Technology 12: 883-888.).

Some strategies have already proved effective in eliminating or reducing such problems, such as the use of “clean” vectors of prokaryotic sequences, absent from obstacles to the coupling of RNA polymerase II and that may lead to the formation of double-stranded mRNA; techniques for integrating single copies of the transgene into the plant genome; the addition of flanking sequences of the scaffold attachment regions and site-directed integration; the choice of germplasm with low methylation activity; in addition to the reduction of negative feedback from promoters by the expression of the target enzyme in a different cell compartment from which its substrate is found (Meyer and Saedler 1996MEYER P and SAEDLER H. 1996. Homology-dependent gene silencing in plants. Annu Rev Plant Physiol Plant Mol Biol 47: 23-48.).

STRATEGIES AT THE POST-TRANSCRIPTIONAL LEVELS

Minimization of post-transcriptional silencing

The processing of primary transcripts is crucial for obtaining high levels of heterologous protein. Although most of plant genetic transformation experiments utilize cDNA-like coding sequences originating from libraries constructed from properly processed mRNAs, the presence of introns in the transgene can significantly increase the stability of mRNAs (Töpfer et al. 1993TÖPFER R, MAAS C, HÖRICKE-GRANDPIERRE C, SCHELL J and STEINBISS H-H 1993. Expression vectors for high-level gene expression in dicotyledonous and monocotyledonous plants. Methods in Enzymology: Academic Press, p. 66-78.). This application has already been proven to increase the expression of endogenous genes in monocots, especially in maize plants (Töpfer et al. 1993TÖPFER R, MAAS C, HÖRICKE-GRANDPIERRE C, SCHELL J and STEINBISS H-H 1993. Expression vectors for high-level gene expression in dicotyledonous and monocotyledonous plants. Methods in Enzymology: Academic Press, p. 66-78.).

Polyadenylation sites also exert great influence on the stability of mRNAs and on the levels of gene expression in plant cells.

Detection and elimination, where possible, of specific recognition sites that contribute to the decay of mRNAs in some terminators may also be used as a tool to avoid post-transcriptional silencing (Töpfer et al. 1993).

Translation optimization

The correct functioning of translation initiation is a limiting factor for the level of accumulation of heterologous proteins. The overlap of the translation initiation site with the Kozak consensus sequence of mRNA is a great optimizer of translation levels, even though there are small structural differences in these sequences in animals and plants, something important when the goal is to express human genes in transgenic plants (Kawaguchi and Bailey-Serres 2005KAWAGUCHI R and BAILEY-SERRES J. 2005. mRNA sequence features that contribute to translational regulation in Arabidopsis. Nucleic Acids Research 33: 955-965.).

The translation rate may also vary depending on the availability of specific amino acid transfer RNAs for certain codons in the plant cell. Optimization of codon usage through site-directed silent mutations or the production of previously modified synthetic coding sequences can be very useful in greatly enhancing the translation of eukaryotic mRNAs (Streatfield 2007).

The leader sequences of different plant transcripts have been shown to influence the increased levels of recombinant protein accumulation in plant bioreactors. They can be adapted on a case-by-case basis for different gene/host plant combinations to maximize translation efficiency (Streatfield 2007).

Post-translational protein stability

The levels of transcription and translation are related to the efficiency of biosynthesis of heterologous proteins. Another variable should be considered for the estimation of the production yield or accumulation of protein biopharmaceuticals: their level of degradation after biosynthesis, that is, their degree of stability (Stoger et al. 2005STOGER E, MA JKC, FISCHER R and CHRISTOU P. 2005. Sowing the seeds of success: pharmaceutical proteins from plants. Current Opin Biotechnol 16: 167-173.).

Molecular strategies that act on protein stability, coupled with the choice of promoters, correspond to the most efficient advances in terms of real increases in yield of recombinant biopharmaceuticals and include two distinct non-exclusive approaches: the use of tissue-specific promoters - mainly those associated with endogenous seed genes - and subcellular targeting of polypeptides (“protein targeting”) (Abiri et al. 2015ABIRI R, VALDIANI A, MAZIAH M, SHAHARUDDIN N, SAHEBI M, BALIA YUSOF ZN, ATABAKI N and TALEI D. 2015. A Critical Review of the Concept of Transgenic Plants: Insights into Pharmaceutical Biotechnology and Molecular Farming. Curr Issues Mol Biol 18: 21-42.).

Tissue-specific promoters are regulatory sequences that restrict gene expression spatially to only one or more parts of the plant and may also indirectly regulate expression at a temporal level when the organ destined for the accumulation of heterologous proteins is associated only with a period of the culture cycle (eg flowers and seeds) (Capell and Christou 2004CAPELL T and CHRISTOU P. 2004. Progress in plant metabolic engineering. Curr Opin Biotechnol 15: 148-154.).

A number of tissue-specific promoters have been extensively characterized, such as those controlling the expression of a maize seed zein, wheat glutenin, rice glutelin and pea seed proteins (Ma et al. 2003) .

The expression of heterologous proteins specifically in seeds implies many advantages naturally provided by such organs. Unlike leaves, seeds are natural storage sites for high concentrations of reserve proteins used in embryo nutrition in the early stages of physiological development of the seedling (Stoger et al. 2005).

These organs present not only a suitable biochemical environment, devoid of phenolic compounds and low concentration of hydrolases, but also presents specialized tissues for the highly stable protein accumulation for long periods of time even at room temperature, which reduces the need for special conditions of storage (Takaiwa et al. 2007TAKAIWA F, TAKAGI H, HIROSE S and WAKASA Y. 2007. Endosperm tissue is good production platform for artificial recombinant proteins in transgenic rice. Plant Biotechnol J 5: 84-92.).

As the costs of processing and purification are inversely proportional to the concentration of the product in relation to the plant biomass, the accumulation of high levels of heterologous proteins in a reduced volume leads to a significant decrease in the production costs of recombinant biopharmaceuticals (Stoger et al. 2002STOGER E, SACK M, PERRIN Y, VAQUERO C, TORRES E, TWYMAN RM, CHRISTOU P and FISCHER R. 2002. Practical considerations for pharmaceutical antibody production in different crop systems. Molecular Breeding 9: 149-158.).

Protein Targeting

Subcellular targeting plays a key role in the levels of heterologous protein accumulation, since the cell compartment in which they are found directly influences the folding, assembling, and post-translational modifications processes, as well as preventing immediate degradation and interference of the polypeptides with cell metabolism - quite frequent events in the cytosol (Fahad et al. 2015FAHAD S et al. 2015. Recent developments in therapeutic protein expression technologies in plants. Biotechnol Lett 37: 265-279.).

N-or C-terminal signal peptides can be fused to heterologous proteins to target specific sites in the cell. These sequences can target proteins to the mitochondria, vacuoles, chloroplasts or retain them in the endoplasmic reticulum, and are generally cleaved after the arrival of the polypeptide of interest to the target organelle (Xu et al. 2012).

Commonly four subcellular targets are the main compartmentalization targets to produce biopharmaceuticals: the apoplast, the endoplasmic reticulum, the chloroplasts and the seed protein bodies (Daniell et al. 2001DANIELL H, STREATFIELD SJ and WYCOFF K. 2001. Medical molecular farming: production of antibodies, biopharmaceuticals and edible vaccines in plants. Trends Plant Sci 6: 219-226.).

Depending on the molecular mass the polypeptide can be secreted or retained in the apoplast, which leads to important applications for suspension cell culture systems. Schillberg et al. (1999)SCHILLBERG S, ZIMMERMANN S, VOSS A and FISCHER R. 1999. Apoplastic and cytosolic expression of full-size antibodies and antibody fragments in Nicotiana tabacum. Transgenic Res 8: 255-263. compared the stability of identical whole antibodies whose accumulation was directed to the cytosol and to the apoplast of transgenic tobacco leaves and found that the secretory pathway constitutes a set of environments more suitable for the folding and assembly of this type of complex protein, since levels of accumulation in the apoplast were much higher than those of the cytosol.

The main disadvantage of addressing to the apoplast is the fact that heterologous proteins are required to be processed before in the Golgi complex, where the addition of typical glycans occurs, which can lead to loss of structural and functional authenticity of the polypeptides (Daniell et al. 2001)

The passage of proteins through the reticulum is the initial destination of polypeptides forwarded to the secretory pathway and the retention of these molecules in the lumen of this organelle has been shown to be more advisable for the accumulation of larger amounts of recombinant proteins in plants than the apoplast itself and the cytosol (Zimmermann et al. 1998ZIMMERMANN S, SCHILLBERG S, LIAO Y-C and FISHER R. 1998. Intracellular expression of TMV-specific single-chain Fv fragments leads to improved virus resistance in shape Nicotiana tabacum. Mol Breed 4(4): 369-379.).

Conrad and Fiedler (1998)CONRAD U and FIEDLER U. 1998. Compartment-specific accumulation of recombinant immunoglobulins in plant cells: an essential tool for antibody production and immunomodulation of physiological functions and pathogen activity. Plant Mol Biol 38: 101-109. determined that the amount of recombinant antibodies fused in the C-terminal portion to the tetrapeptide K/H DEL - one of the most popular peptide-signals currently employed for the retention of heterologous proteins in the lumen of the endoplasmic reticulum - was 2 a 10 times greater than those expressed in different plant species in the absence of the signal peptide.

The use of K/H DEL also accounted for a marked increase in the accumulation of DIP B protein in leaves cells of Arabidopsis thaliana, resulting in 8.5% of total soluble proteins in these organs (Yang et al. 2005YANG J, BARR LA, FAHNESTOCK SR and LIU Z-B. 2005. High yield recombinant silk-like protein production in transgenic plants through protein targeting. Transgenic Res 14: 313-324.).

Polypeptides expressed in cereal seeds remained stable up to three years after storage at room temperature, with no significant loss of activity due to the low proteolytic activity in these compartments (Larrick and Thomas 2001LARRICK JW and THOMAS DW. 2001. Producing proteins in transgenic plants and animals. Curr Opin Biotechnol 12: 411-418.).

Another advantage of so-called protein targeting for the endoplasmic reticulum of seeds is the fact that proteins retained in this organelle do not undergo processing in the Golgi complex, a fundamental aspect for the production of heterologous proteins and recombinant antibodies in particular (Stoger et al. 2002).

Protein targeting for chloroplasts is also an interesting strategy to increase levels of recombinant protein accumulation. In the chloroplast lumen there are chaperones capable of assisting in the folding and assembly of heterodimeric and oligomeric proteins, in addition to low concentrations of hydrolases and peptidases, which makes this environment relatively stable for the accumulation of proteins of interest (Daniell et al. 2001).

However, since the machinery of gene expression in chloroplasts resembles that of prokaryotes, most post-translational modifications, such as glycosylation, cannot be performed on this organelle, except for the formation of disulfide bonds (Tschofen et al. 2016).

Protein bodies, in turn, are extensions derived from the endoplasmic reticulum specialized in the accumulation of reserve proteins in seeds of several species of grain-producing plants. They function as cisterns that occupy a large cytoplasmic volume and evolutionarily have undergone adaptations resulting in the high capacity to compartmentalize large volumes of proteins and to maximize the integrity of these polypeptides (Zheng et al. 1992ZHENG Y, HE M, HAO S and HUANG B. 1992. The Ultrastructural Evidence on the Origin of Protein Bodies in the Rough Endoplasmic Reticulum of Developing Cotyledons of Soybean. Ann Bot 69(5): 377-383.).

These globular inclusions are found in cotyledonary cells of tobacco seeds, legumes such as soybeans and beans, as well as grasses such as wheat and barley, and originate now when biosynthesis of reserve proteins begins in the tissues responsible for nourishing the embryo (Yoo and Chrispeels 1980YOO BY and CHRISPEELS MJ. 1980. The origin of protein bodies in developing soybean cotyledons: a proposal. Protoplasma 103: 201-204.). In this way there is a significant increase in the number and availability of protein bodies throughout the process of physiological maturation of the grains of these species.

Since these highly specialized vacuoles do not undergo any type of fusion with lysosomes, their lumen presents near-neutral pH and practically the absence of aminopeptidases, factors that characterize them as a subcellular environment where protein degradation is minimal and an excellent target for the addressing heterologous polypeptides when the main goal of genetic transformation is to maintain the stability of these molecules (Takaiwa et al. 2007).

MAGNIFECTIONTM: A METHOD FOR THE MASS EXPRESSION OF HETEROLOGOUS PROTEINS AND PEPTIDES IN NICOTIANA BENTHAMIANA

As responsive elements of innate defense systems of microbes, plants and animals, AMPs are naturally synthesized at low levels (Watkins and Bonomo 2016WATKINS RR and BONOMO RA. 2016. Overview: Global and Local Impact of Antibiotic Resistance. Infect Dis Clin North Am 30: 313-322., Uhlig et al. 2014UHLIG T, KYPRIANOU T, MARTINELLI FG, OPPICI CA, HEILIGERS D, HILLS D, CALVO XR and VERHAERT P. 2014. The emergence of peptides in the pharmaceutical business: From exploration to exploitation. EuPA Open Proteomics 4: 58-69.). To maximize peptide biosynthesis, genetic engineered bacteria and yeast cells are frequently explored as vehicles for the recombinant production of bioactive AMPs (Perry et al. 2016PERRY J, WAGLECHNER N and WRIGHT G. 2016. The Prehistory of Antibiotic Resistance. Cold Spring Harb Perspect Med 2016: 6(6)., Nagel et al. 2016NAGEL TE, CHAN BK, DE VOS D, EL-SHIBINY A, KANG’ETHE EK, MAKUMI A and PIRNAY J-P. 2016. The Developing World Urgently Needs Phages to Combat Pathogenic Bacteria. Front Microbiol 7: 882.). To this day, many different AMPs have been synthesized in E. coli and Pichia pastoris (Perry et al. 2016PERRY J, WAGLECHNER N and WRIGHT G. 2016. The Prehistory of Antibiotic Resistance. Cold Spring Harb Perspect Med 2016: 6(6).). Apart from the high therapeutic potential presented by recombinant AMPs, limited investment of companies and drawbacks in terms of poor yield, low quality and unsatisfactory in vivo activity restricted the commercial development to few promising AMPs. Besides production limitations, some of them reached advanced clinical trials prior to commercialization.

Among the most important factors that limit the recombinant production of AMPs in microbial systems are the inner toxicity of the peptide for the host cells – for many AMPs, even low concentrations can be fatal for the microbe – and low quality of product in terms of post-translational modifications. Under such circumstances, plant appear as interesting alternative systems for the production of recombinant AMPs (Kosikowska and Lesner 2016KOSIKOWSKA P and LESNER A. 2016. Antimicrobial peptides (AMPs) as drug candidates: a patent review (2003–2015). Expert Opin Ther Pat 26: 689-702., Nagel et al. 2016NAGEL TE, CHAN BK, DE VOS D, EL-SHIBINY A, KANG’ETHE EK, MAKUMI A and PIRNAY J-P. 2016. The Developing World Urgently Needs Phages to Combat Pathogenic Bacteria. Front Microbiol 7: 882.).

Although plants perform a vast arsenal of post-translational modifications, low levels of recombinant biosynthesis of peptides are common, resulting in low quantities of purified products. In addition to such limitations, plant systems for expression of heterologous proteins have undergone enormous technical improvement in recent years. The discovery of strong promoters associated with increased gene expression, strategies and elements that stabilize primary transcripts, and optimization of codons and methods that maximize posttranslational stability have contributed to dramatic increases in the final amounts of proteins accumulated in transgenic plants and In suspension of plant cells (Obembe et al. 2011). Despite significant improvements, limitations such as the usually low levels of expression and the long time to obtain enough protein quantities for the first biological assays still restrict recombinant production in these systems (Xu et al. 2012).

The most interesting and recent alternative to circumvent both limitations is the use of viral vectors of gene carrier plants of interest that provide high levels of transient expression within a few days after inoculation. The MagnifectionTM system integrates methods for expressing transgenes of interest using viral vectors specially constructed to provide high transcription rates, high speed in obtaining the first milligrams of heterologous protein and high scalability (Gleba et al. 2005GLEBA Y, KLIMYUK V and MARILLONNET S. 2005. Magnifection—a new platform for expressing recombinant vaccines in plants. Vaccine 23: 2042-2048.).

In essence, the the MagnifectionTM strategy advocates total infiltration of adult plants of N. benthamiana with a diluted suspension of Agrobacterium tumefaciens containing RNA replicon encoding T-DNAs, usually based on tobacco mosaic virus (TMV). In this process, the bacterium assumes the initial viral functions of primary infection and systemic movement, while the viral vector promotes short scattering (cell to cell), signal amplification, and expression of the transgene of interest at high levels. Thus, few adult tobacco plants allow the preliminary optimization of dilutions of the suspensions and the obtaining of milligrams or grams of the heterologous protein (Gleba et al. 2014) (Figure 1).

Figure 1
Agroinfiltration method for the large-scale biosynthesis of recombinant AMPs. After gene cloning using E. coli cells, pro-vectors carrying the 3`extremity of the expression cassette (green vector), the 5`extremity of the expression cassette (blue vector) and the gene that codifies the integrase for the extremities recombination (yellow vector) are inserted in A. tumefaciens cells, followed by cultivation in LB medium. The bacterial culture is then injected in the downside of N. benthamiana leaves. After 5 to 8 days, yields of recombinant AMPs can be obtained from harvested leaves after extraction and purification.

The MagnifectionTM technology combines the advantages of standard expression systems (viral, bacterial and plant) and presents three major advantages when compared to other platforms. The first one is the use of efficient viral vectors with a design that allows high gene expression. This allows high rates of transgene expression, especially with respect to the increase of gene transcription, guaranteed by the presence of strong viral promoters. The presence of introns in the expression cassette also allow for the correct migration of the mRNA into the cytosol of the plant cell for the massive production of recombinant proteins (Gleba et al. 2004GLEBA Y, MARILLONNET S and KLIMYUK V. 2004. Engineering viral expression vectors for plants: the ‘full virus’ and the ‘deconstructed virus’ strategies. Curr Opin Plant Biol 7: 182-188., 2014GLEBA YY, TUSÉ D and GIRITCH A 2014. Plant Viral Vectors for Delivery by Agrobacterium. In: Palme, K and Gleba Y (Eds), Plant Viral Vectors, Berlin, Heidelberg: Springer Berlin Heidelberg, p. 155-192.).

Secondly, Agrobacterium promotes the genetic transformation of somatic cells of N. benthamiana leaves in a systemic way. With cell-to-cell bacterial turnover, there is no need to increase the viral expression cassette with superfluous genes, such as viral motility coding genes, for example. This increases the spread of transgenes integrated into the plant chromosome and does not allow metabolic shifts to the biosynthesis of additional proteins without utility, concentrating the entire production effort on the transcription and translation machinery (Gleba et al. 2004GLEBA Y, MARILLONNET S and KLIMYUK V. 2004. Engineering viral expression vectors for plants: the ‘full virus’ and the ‘deconstructed virus’ strategies. Curr Opin Plant Biol 7: 182-188.,2007GLEBA Y, KLIMYUK V and MARILLONNET S. 2007. Viral vectors for the expression of proteins in plants. Curr Opin Biotechnol 18: 134-141.).

Finally, the systems allow unprecedented speed ​​and efficiency of accumulation of transcripts equal to those of a plant retrovirus and consistently high production yields at low cost (Gleba et al. 2005GLEBA Y, KLIMYUK V and MARILLONNET S. 2005. Magnifection—a new platform for expressing recombinant vaccines in plants. Vaccine 23: 2042-2048.). The high scalability of agroinfection of plants with transgenic A. tumefaciens is due to the high inoculation yield – 1 litre of bacterial culture medium is sufficient for the infection of more than 10 kg of leaves - and to the rapid peak of gene expression, which normally occurs between the seventh and the eighth days after the agroinfiltration (Figure 2).

Figure 2
Expression kinetics of foreign genes in N. benthamiana leaves. The gene delivery strategy mediated by A. tumefaciens allows the efficient spread of AMPs coding genes and integration to the host cells. Potent viral promoters induce high expression rates of the Green Fluorescent Protein (GFP) from Aequorea victoria gene, a frequent reporter gene used for experimental validation. The peak of expression is about the seventh or eighth day after inoculation.

In a synthetic way, the MagnifectionTM system allows the industrial production of proteins and peptides without the genetic transformation of germ cells, which leads to rapid and safe protein synthesis.

As an alternative to the stable transformation of nuclear and plastid genomes, transient expression consists of the transient transformation of somatic cells by genes systemically carried by Agrobacterium and in the amplification of protein accumulation based on viral vectors in leaf cells. The MagnifectionTM system uses the strategy of “deconstructed” viral vectors, i.e., vectors containing lean expression cassettes that dispense superfluous elements related to protein translation, the assembly of mature virions and their spread over long distances, the reprogramming of metabolism Host plant, inhibition of silencing and etc. Many of these actions are delegated to Agrobacterium, notably spreading, the host plant (Marillonnet et al. 2004MARILLONNET S, GIRITCH A, GILS M, KANDZIA R, KLIMYUK V and GLEBA Y. 2004. In planta engineering of viral RNA replicons: Efficient assembly by recombination of DNA modules delivered by Agrobacterium. Proc Natl Acad Sci U S A 101: 6852-6857., 2005).

The industrial scale can be easily obtained by means of a simple apparatus for vacuum agroinfiltration of whole plants, wherein the incubation time for viral amplification and peak expression does not exceed 10 days. Frequently, 5 grams of heterologous proteins are obtained per kilogram of infiltrated dry matter, and levels of expression equal to 80% of total soluble proteins are common in greenhouse (Gleba et al. 2004).

Since 2010, the MagnifectionTM system has been used by the Canadian company Medicago for the industrial production of a vaccine against influenza caused by the H1N1 virus in the United States. A $ 21 million funding agreement was signed between Medicago and the American Defense Advanced Agency Agency (DARPA) to produce 10 million doses / month in the event of a pandemic. The vaccine is undergoing phase II clinical trials (in a total of IV) and tends to join another vaccine against the viral variant H5N1, produced with the same system (Holtz et al. 2015HOLTZ BR, BERQUIST BR, BENNETT LD, KOMMINENI VJM, MUNIGUNTI RK, WHITE EL, WILKERSON DC, WONG KYI, LY LH and MARCEL S. 2015. Commercial-scale biotherapeutics manufacturing facility for plant-made pharmaceuticals. Plant Biotechnol J 13: 1180-1190.).

Today, many antigens have already been synthesized using the MagnifectionTM system, as shown in Table IV.

TABLE IV
Examples of transiently expressed antigens using the MagnifectionTM system.

A series of modern viral vectors based on TMV, called PICHs, are optimized for the expression of pharmacological proteins. Such plasmids, referred to as pro-vectors, contain tobacco introns systematically arranged throughout the molecule for processing in the nucleus and recombination sites that allow manipulation of the DNA still within the cell prior to conversion into a hybrid amplicon of RNA. DNA molecules containing the genes of interest can be efficiently recombined in plant, using site-specific recombinases such as Cre or the Integrase C31 from Streptomyces sp. (Gleba et al. 2014). To obtain an RNA without the recombination sites, engineered as introns by the RNA processing machinery of the nucleus. In summary, the process advocates the in vivo assembly of DNA pro-vectors by simple mixing and co-infiltration of plants with Agrobacterium cells that carry the components of the vector separately (Figure 3).

Figure 3
Pro-vector system for assembly of gene expression modules. Two viral vectors, donors respectively of the 5 ‘and 3’ ends, are specifically recognized by the enzyme Integrase C31 from Streptomyces sp. The enzyme catalyzes the recombination between the fragments by the pairing of homologous bases at the recombination sites. The final hybrid fragment presents all genetic elements for large-scale gene expression and for the addressing / purification of recombinant AMP.

In this way, a series of three pro-vectors must be used simultaneously in the experiments to obtain high levels of recombinant expression. This approach allows rapid assembly and expression of protein variants by the combination of peptide signals, facilitating purification tails, binding domains and transit peptides, in a versatile arrangement that provides milligrams of proteins in only a few days (Gleba et al. 2004GLEBA Y, MARILLONNET S and KLIMYUK V. 2004. Engineering viral expression vectors for plants: the ‘full virus’ and the ‘deconstructed virus’ strategies. Curr Opin Plant Biol 7: 182-188., 2014, Marillonnet et al. 2004, 2005MARILLONNET S, THOERINGER C, KANDZIA R, KLIMYUK V and GLEBA Y. 2005. Systemic Agrobacterium tumefaciens-mediated transfection of viral replicons for efficient transient expression in plants. Nat Biotech 23: 718-723.). Due to all the properties exposed, the MagnifectionTM system can be an excellent platform for the large-scale biosynthesis of the antimicrobial peptides Cn-AMP1, Clavanin A, Cm AMP-5 and Parigidin-br1.

CONCLUSIONS

Recombinant expression of the AMPs using the MagnifectionTM system will provide subsidies for the systematization of large-scale expression of molecules with very high therapeutic potential and ability to inhibit a wide range of pathogens that threaten human health. Pro-vector combinations represent a great advance concerning the production of recombinant therapeutic molecules. As a consequence of their self-assembly ability, viral vectors may be used to study the expression of several other AMPs extracted from other plants, for example, thionines, snakines, heveins, defensins, lipid-transferring proteins and other cytotoxic agents. Such improvements allow the production of large quantities of AMPs sufficient to test new antimicrobial control functions with potential application in agriculture, pharmacology and food processing.

The massive biosynthesis of AMPs transiently accumulated in N. benthamiana leaves has great scientific impact, since it creates conditions for the systematic study of the rapid and mass expression of antimicrobial peptides in a cheap and safe system.

In addition, the industrial scale production of AMPs in a simple recombinant system with high capacity of production scheduling, besides the cultivation of greenhouse plants under conventional growth conditions, with only water, light and fertile soil, represents a great economic potential for the generation of efficient and cheap products with high value.

It is worth mentioning that this transient expression approach using deconstructed viral pro-vectors is a pioneer technology that represents the cutting edge of what is most sophisticated in terms of the use of heterologous vegetative systems for therapeutic purposes.

ACKNOWLEGMENTS

We thank the Academia Brasileira de Ciências for the invitation to write this article. This work was funded by the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Fundação de Apoio à Pesquisa do Distrito Federal (FAPDF) and Fundação de Apoio ao Desenvolvimento do Ensino, Ciência e Tecnologia do Estado de Mato Grosso do Sul (FUNDECT).

REFERENCES

  • ABIRI R, VALDIANI A, MAZIAH M, SHAHARUDDIN N, SAHEBI M, BALIA YUSOF ZN, ATABAKI N and TALEI D. 2015. A Critical Review of the Concept of Transgenic Plants: Insights into Pharmaceutical Biotechnology and Molecular Farming. Curr Issues Mol Biol 18: 21-42.
  • ANDERSSON DI, HUGHES D and KUBICEK-SUTHERLAND JZ. 2016. Mechanisms and consequences of bacterial resistance to antimicrobial peptides. Drug Resist Updat 26: 43-57.
  • CAPELL T and CHRISTOU P. 2004. Progress in plant metabolic engineering. Curr Opin Biotechnol 15: 148-154.
  • CHEN GH, HSU MP, TAN CH, SUNG HY, KUO CG, FAN MJ, CHEN HM, CHEN S and CHEN CS. 2005. Cloning and Characterization of a Plant Defensin VaD1 from Azuki Bean. J Agric Food Chem 53: 982-988.
  • CONRAD U and FIEDLER U. 1998. Compartment-specific accumulation of recombinant immunoglobulins in plant cells: an essential tool for antibody production and immunomodulation of physiological functions and pathogen activity. Plant Mol Biol 38: 101-109.
  • CRAIK DJ, ČEMAŽAR M, WANG CKL and DALY NL. 2006. The cyclotide family of circular miniproteins: Nature’s combinatorial peptide template. J Pept Sci 84: 250-266.
  • CUNHA NB et al. 2017. The next generation of antimicrobial peptides (AMPs) as molecular therapeutic tools for the treatment of diseases with social and economic impacts. Drug Discov Today 22: 234-248.
  • CUNHA NB et al. 2011a. Expression of functional recombinant human growth hormone in transgenic soybean seeds. Transgenic Res 20: 811-826.
  • CUNHA NB et al. 2011b. Accumulation of functional recombinant human coagulation factor IX in transgenic soybean seeds. Transgenic Res 20: 841-855.
  • CUNHA NBD et al. 2016. Cloning and characterization of novel cyclotides genes from South American plants. J Pept Sci 106: 784-795.
  • DA CUNHA NB, VIANNA GR, DA ALMEIDA LIMA T and RECH E. 2014. Molecular farming of human cytokines and blood products from plants: Challenges in biosynthesis and detection of plant-produced recombinant proteins. Biotechnol J 9: 39-50.
  • DANIELL H, STREATFIELD SJ and WYCOFF K. 2001. Medical molecular farming: production of antibodies, biopharmaceuticals and edible vaccines in plants. Trends Plant Sci 6: 219-226.
  • DAVIES HM. 2010. Commercialization of whole-plant systems for biomanufacturing of protein products: evolution and prospects. Plant Biotechnol J 8: 845-861.
  • DEL PRETE G, SANTI L, ANDRIANAIVOARIMANANA V, AMEDEI A, DOMARLE O, D’ ELIOS MM, ARNTZEN CJ, RAHALISON L and MASON HS. 2009. Plant-derived recombinant F1, V, and F1-V fusion antigens of Yersinia pestis activate human cells of the innate and adaptive immune system. Int J Immunopathol Pharmacol 22: 133-143.
  • DRAKE PMW, SZETO TH, PAUL MJ, TEH AYH and MA JKC. 2017. Recombinant biologic products versus nutraceuticals from plants – a regulatory choice? Br J Clin Pharmacol 83: 82-87.
  • EGELKROUT E, RAJAN V and HOWARD JA. 2012. Overproduction of recombinant proteins in plants. Plant Sci 184: 83-101.
  • FAHAD S et al. 2015. Recent developments in therapeutic protein expression technologies in plants. Biotechnol Lett 37: 265-279.
  • FINNEGAN J and MCELROY D. 1994. Transgene Inactivation: Plants Fight Back! Bio/Technology 12: 883-888.
  • FISCHER R, STOGER E, SCHILLBERG S, CHRISTOU P and TWYMAN RM. 2004. Plant-based production of biopharmaceuticals. Curr Opin Plant Biol 7: 152-158.
  • FRANCONI R, DEMURTAS OC and MASSA S. 2010. Plant-derived vaccines and other therapeutics produced in contained systems. Expert Rev Vaccines 9: 877-892.
  • GLEBA Y, KLIMYUK V and MARILLONNET S. 2005. Magnifection—a new platform for expressing recombinant vaccines in plants. Vaccine 23: 2042-2048.
  • GLEBA Y, KLIMYUK V and MARILLONNET S. 2007. Viral vectors for the expression of proteins in plants. Curr Opin Biotechnol 18: 134-141.
  • GLEBA Y, MARILLONNET S and KLIMYUK V. 2004. Engineering viral expression vectors for plants: the ‘full virus’ and the ‘deconstructed virus’ strategies. Curr Opin Plant Biol 7: 182-188.
  • GLEBA YY, TUSÉ D and GIRITCH A 2014. Plant Viral Vectors for Delivery by Agrobacterium. In: Palme, K and Gleba Y (Eds), Plant Viral Vectors, Berlin, Heidelberg: Springer Berlin Heidelberg, p. 155-192.
  • GU Q, HAN N, LIU J and ZHU M. 2005. Cloning of Helicobacter pylori urease subunit B gene and its expression in tobacco (Nicotiana tabacum L.). Plant Cell Rep 24: 532-539.
  • HOLTZ BR, BERQUIST BR, BENNETT LD, KOMMINENI VJM, MUNIGUNTI RK, WHITE EL, WILKERSON DC, WONG KYI, LY LH and MARCEL S. 2015. Commercial-scale biotherapeutics manufacturing facility for plant-made pharmaceuticals. Plant Biotechnol J 13: 1180-1190.
  • KAWAGUCHI R and BAILEY-SERRES J. 2005. mRNA sequence features that contribute to translational regulation in Arabidopsis. Nucleic Acids Research 33: 955-965.
  • KIM KY, KWON SY, LEE HS, HUR Y, BANG JW and KWAK SS. 2003. A novel oxidative stress-inducible peroxidase promoter from sweetpotato: molecular cloning and characterization in transgenic tobacco plants and cultured cells. Plant Mol Biol 51: 831-838.
  • KOSIKOWSKA P and LESNER A. 2016. Antimicrobial peptides (AMPs) as drug candidates: a patent review (2003–2015). Expert Opin Ther Pat 26: 689-702.
  • KOYA V, MOAYERI M, LEPPLA SH and DANIELL H. 2005. Plant-Based Vaccine: Mice Immunized with Chloroplast-Derived Anthrax Protective Antigen Survive Anthrax Lethal Toxin Challenge. Infect Immun 73: 8266-8274.
  • LARRICK JW and THOMAS DW. 2001. Producing proteins in transgenic plants and animals. Curr Opin Biotechnol 12: 411-418.
  • LEE MYT, ZHOU Y, LUNG RWM, CHYE M-L, YIP W-K, ZEE S-Y and LAM E. 2006. Expression of viral capsid protein antigen against Epstein-Barr virus in plastids of Nicotiana tabacum cv. SR1. Biotechnol Bioeng 94: 1129-1137.
  • LENZI P et al. 2008. Translational fusion of chloroplast-expressed human papillomavirus type 16 L1 capsid protein enhances antigen accumulation in transplastomic tobacco. Transgenic Res 17: 1091-1102.
  • LIENARD D et al. 2007. Suspension-cultured BY-2 tobacco cells produce and mature immunologically active house dust mite allergens. Plant Biotechnol J 5: 93-108.
  • LOH H-S, GREEN BJ and YUSIBOV V. 2017. Using transgenic plants and modified plant viruses for the development of treatments for human diseases. Curr Opin in Virol 26: 81-89.
  • LÓPEZ-ABARRATEGUI C et al. 2015. Cm-p5: an antifungal hydrophilic peptide derived from the coastal mollusk Cenchritis muricatus (Gastropoda: Littorinidae). FASEB J 29: 3315-3325.
  • MA JKC, CHIKWAMBA R, SPARROW P, FISCHER R, MAHONEY R and TWYMAN RM. 2005. Plant-derived pharmaceuticals - the road forward. Trends Plant Sci 10: 580-585.
  • MA JKC, DRAKE PMW and CHRISTOU P. 2003. The production of recombinant pharmaceutical proteins in plants. Nat Rev Genet 4: 794-805.
  • MA S, HUANG Y, YIN Z, MENASSA R, BRANDLE JE and JEVNIKAR AM. 2004. Induction of oral tolerance to prevent diabetes with transgenic plants requires glutamic acid decarboxylase (GAD) and IL-4. Proc Natl Acad Sci U S A 101: 5680-5685.
  • MALANOVIC N and LOHNER K. 2016. Gram-positive bacterial cell envelopes: The impact on the activity of antimicrobial peptides. Biochim Biophys Acta 1858: 936-946.
  • MANDAL SM, DEY S, MANDAL M, SARKAR S, MARIA-NETO S and FRANCO OL. 2009. Identification and structural insights of three novel antimicrobial peptides isolated from green coconut water. Peptides 30: 633-637.
  • MARILLONNET S, GIRITCH A, GILS M, KANDZIA R, KLIMYUK V and GLEBA Y. 2004. In planta engineering of viral RNA replicons: Efficient assembly by recombination of DNA modules delivered by Agrobacterium. Proc Natl Acad Sci U S A 101: 6852-6857.
  • MARILLONNET S, THOERINGER C, KANDZIA R, KLIMYUK V and GLEBA Y. 2005. Systemic Agrobacterium tumefaciens-mediated transfection of viral replicons for efficient transient expression in plants. Nat Biotech 23: 718-723.
  • MARSIAN J and LOMONOSSOFF GP. 2016. Molecular pharming—VLPs made in plants. Curr Opin Biotechnol 37: 201-206.
  • MCCORMICK AA et al. 2008. Plant-produced idiotype vaccines for the treatment of non-Hodgkin’s lymphoma: Safety and immunogenicity in a phase I clinical study. Proc Natl Acad Sci U S A 105: 10131-10136.
  • MEYER P and SAEDLER H. 1996. Homology-dependent gene silencing in plants. Annu Rev Plant Physiol Plant Mol Biol 47: 23-48.
  • MEYERS B, ZALTSMAN A, LACROIX B, KOZLOVSKY SV and KRICHEVSKY A. 2010. Nuclear and plastid genetic engineering of plants: Comparison of opportunities and challenges. Biotechnol Adv 28: 747-756.
  • MOJSOSKA B and JENSSEN H. 2015. Peptides and Peptidomimetics for Antimicrobial Drug Design. Pharmaceuticals 8: 366-415.
  • NAGEL TE, CHAN BK, DE VOS D, EL-SHIBINY A, KANG’ETHE EK, MAKUMI A and PIRNAY J-P. 2016. The Developing World Urgently Needs Phages to Combat Pathogenic Bacteria. Front Microbiol 7: 882.
  • NGUYEN GKT, LIM WH, NGUYEN PQT and TAM JP. 2012. Novel Cyclotides and Uncyclotides with Highly Shortened Precursors from Chassalia chartacea and Effects of Methionine Oxidation on Bioactivities. J Biol Chem 287: 17598-17607.
  • NGUYEN LT, HANEY EF and VOGEL HJ. 2011. The expanding scope of antimicrobial peptide structures and their modes of action. Trends Biotechnol 29: 464-472.
  • OBEMBE OO, POPOOLA JO, LEELAVATHI S and REDDY SV. 2011. Advances in plant molecular farming. Biotechnol Adv 29: 210-222.
  • PADIDAM M. 2003. Chemically regulated gene expression in plants. Curr Opin Plant Biol 6: 169-177.
  • PARACHIN NS and FRANCO OL. 2014. New edge of antibiotic development: antimicrobial peptides and corresponding resistance. Front Microbiol 5: 147.
  • PARACHIN NS, MULDER KC, VIANA AA, DIAS SC and FRANCO OL. 2012. Expression systems for heterologous production of antimicrobial peptides. Peptides:38(2): 446-456.
  • PELEGRINI PB, QUIRINO BF and FRANCO OL. 2007. Plant cyclotides: An unusual class of defense compounds. Peptides 28: 1475-1481.
  • PERRY J, WAGLECHNER N and WRIGHT G. 2016. The Prehistory of Antibiotic Resistance. Cold Spring Harb Perspect Med 2016: 6(6).
  • PINTO MFS et al. 2016. Characterization of a Bioactive Acyclotide from Palicourea rigida. J Nat Prod 79: 2767-2773.
  • POGREBNYAK N, GOLOVKIN M, ANDRIANOV V, SPITSIN S, SMIRNOV Y, EGOLF R and KOPROWSKI H. 2005. Severe acute respiratory syndrome (SARS) S protein production in plants: Development of recombinant vaccine. Proc Natl Acad Sci U S A 102: 9062-9067.
  • REGNARD GL, HALLEY-STOTT RP, TANZER FL, HITZEROTH II and RYBICKI EP. 2010. High level protein expression in plants through the use of a novel autonomously replicating geminivirus shuttle vector. Plant Biotechnol J 8: 38-46.
  • SANTANA MJ, DE OLIVEIRA AL, QUEIROZ JÚNIOR LHK, MANDAL SM, MATOS CO, DE O. DIAS R, FRANCO OL and LIÃO LM. 2015. Structural insights into Cn-AMP1, a short disulfide-free multifunctional peptide from green coconut water. FEBS Lett 589: 639-644.
  • SCHILLBERG S, ZIMMERMANN S, VOSS A and FISCHER R. 1999. Apoplastic and cytosolic expression of full-size antibodies and antibody fragments in Nicotiana tabacum. Transgenic Res 8: 255-263.
  • SHAALTIEL Y, GINGIS–VELITSKI S, TZABAN S, FIKS N, TEKOAH Y and AVIEZER D. 2015. Plant-based oral delivery of β-glucocerebrosidase as an enzyme replacement therapy for Gaucher’s disease. Plant Biotechnol J 13: 1033-1040.
  • SILVA ON, FENSTERSEIFER ICM, RODRIGUES EA, HOLANDA HHS, NOVAES NRF and CUNHA JPA. 2015. Clavanin A improves outcome of complications from different bacterial infections. Antimicrob Agents Chemother 59(3): 1620-1626.
  • SILVA ON, MULDER KC, BARBOSA AE, OTERO-GONZALEZ AJ, LOPEZ-ABARRATEGUI C and REZENDE TM. 2011. Exploring the pharmacological potential of promiscuous host-defense peptides: from natural screenings to biotechnological applications. Front Microbiol 22: 2;232.
  • SILVA ON et al. 2012. Cn-AMP1: A new promiscuous peptide with potential for microbial infections treatment. Pept Sci 98: 322-331.
  • STOGER E, MA JKC, FISCHER R and CHRISTOU P. 2005. Sowing the seeds of success: pharmaceutical proteins from plants. Current Opin Biotechnol 16: 167-173.
  • STOGER E, SACK M, PERRIN Y, VAQUERO C, TORRES E, TWYMAN RM, CHRISTOU P and FISCHER R. 2002. Practical considerations for pharmaceutical antibody production in different crop systems. Molecular Breeding 9: 149-158.
  • STREATFIELD SJ. 2007. Approaches to achieve high-level heterologous protein production in plants. Plant Biotechnol J 5: 2-15.
  • TAKAIWA F, TAKAGI H, HIROSE S and WAKASA Y. 2007. Endosperm tissue is good production platform for artificial recombinant proteins in transgenic rice. Plant Biotechnol J 5: 84-92.
  • THANAVALA Y, YANG YF, LYONS P, MASON HS and ARNTZEN C. 1995. Immunogenicity of transgenic plant-derived hepatitis B surface antigen. Proc Natl Acad Sci U S A 92: 3358-3361.
  • TÖPFER R, MAAS C, HÖRICKE-GRANDPIERRE C, SCHELL J and STEINBISS H-H 1993. Expression vectors for high-level gene expression in dicotyledonous and monocotyledonous plants. Methods in Enzymology: Academic Press, p. 66-78.
  • TREGONING JS, CLARE S, BOWE F, EDWARDS L, FAIRWEATHER N, QAZI O, NIXON PJ, MALIGA P, DOUGAN G and HUSSELL T. 2005. Protection against tetanus toxin using a plant-based vaccine. Eur J Immunol 35: 1320-1326.
  • TSCHOFEN M, KNOPP D, HOOD E and STÖGER E. 2016. Plant Molecular Farming: Much More than Medicines. Annu Rev Analyt Chem 9: 271-294.
  • TWYMAN RM, STOGER E, SCHILLBERG S, CHRISTOU P and FISCHER R. 2003. Molecular farming in plants: host systems and expression technology. Trends Biotechnol 21: 570-578.
  • UHLIG T, KYPRIANOU T, MARTINELLI FG, OPPICI CA, HEILIGERS D, HILLS D, CALVO XR and VERHAERT P. 2014. The emergence of peptides in the pharmaceutical business: From exploration to exploitation. EuPA Open Proteomics 4: 58-69.
  • WATKINS RR and BONOMO RA. 2016. Overview: Global and Local Impact of Antibiotic Resistance. Infect Dis Clin North Am 30: 313-322.
  • WU L, JIANG L, ZHOU Z, FAN J, ZHANG Q, ZHU H, HAN Q and XU Z. 2003. Expression of foot-and-mouth disease virus epitopes in tobacco by a tobacco mosaic virus-based vector. Vaccine 21: 4390-4398.
  • XU J, DOLAN MC, MEDRANO G, CRAMER CL and WEATHERS PJ. 2012. Green factory: Plants as bioproduction platforms for recombinant proteins. Biotechnol Adv 30: 1171-1184.
  • YANG J, BARR LA, FAHNESTOCK SR and LIU Z-B. 2005. High yield recombinant silk-like protein production in transgenic plants through protein targeting. Transgenic Res 14: 313-324.
  • YOO BY and CHRISPEELS MJ. 1980. The origin of protein bodies in developing soybean cotyledons: a proposal. Protoplasma 103: 201-204.
  • ZHANG GG, RODRIGUES L, ROVINSKI B and WHITE KA. 2002. Production of HIV-1 p24 protein in transgenic tobacco plants. Mol Biotechnol 20: 131-136.
  • ZHENG Y, HE M, HAO S and HUANG B. 1992. The Ultrastructural Evidence on the Origin of Protein Bodies in the Rough Endoplasmic Reticulum of Developing Cotyledons of Soybean. Ann Bot 69(5): 377-383.
  • ZIMMERMANN S, SCHILLBERG S, LIAO Y-C and FISHER R. 1998. Intracellular expression of TMV-specific single-chain Fv fragments leads to improved virus resistance in shape Nicotiana tabacum. Mol Breed 4(4): 369-379.

Publication Dates

  • Publication in this collection
    18 Oct 2018
  • Date of issue
    2019

History

  • Received
    6 Feb 2018
  • Accepted
    7 June 2018
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br