Next Article in Journal
Ginseng Prong Added to Broiler Diets Reduces Lipid Peroxidation in Refrigerated and Frozen Stored Poultry Meats
Previous Article in Journal
Valorization and Application of Fruit and Vegetable Wastes and By-Products for Food Packaging Materials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biotechnological Insights on the Expression and Production of Antimicrobial Peptides in Plants

by
Balamurugan Shanmugaraj
1,
Christine Joy I. Bulaon
2,3,†,
Ashwini Malla
1,† and
Waranyoo Phoolcharoen
2,3,*
1
Baiya Phytopharm Co., Ltd., Bangkok 10250, Thailand
2
Research Unit for Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
3
Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2021, 26(13), 4032; https://doi.org/10.3390/molecules26134032
Submission received: 7 June 2021 / Revised: 27 June 2021 / Accepted: 28 June 2021 / Published: 1 July 2021

Abstract

:
The emergence of drug-resistant pathogens poses a serious critical threat to global public health and requires immediate action. Antimicrobial peptides (AMPs) are a class of short peptides ubiquitously found in all living forms, including plants, insects, mammals, microorganisms and play a significant role in host innate immune system. These peptides are considered as promising candidates to treat microbial infections due to its distinct advantages over conventional antibiotics. Given their potent broad spectrum of antimicrobial action, several AMPs are currently being evaluated in preclinical/clinical trials. However, large quantities of highly purified AMPs are vital for basic research and clinical settings which is still a major bottleneck hindering its application. This can be overcome by genetic engineering approaches to produce sufficient amount of diverse peptides in heterologous host systems. Recently plants are considered as potential alternatives to conventional protein production systems such as microbial and mammalian platforms due to their unique advantages such as rapidity, scalability and safety. In addition, AMPs can also be utilized for development of novel approaches for plant protection thereby increasing the crop yield. Hence, in order to provide a spotlight for the expression of AMP in plants for both clinical or agricultural use, the present review presents the importance of AMPs and efforts aimed at producing recombinant AMPs in plants for molecular farming and plant protection so far.

1. Introduction

Various antibiotics are used for the prevention or treatment of many common diseases caused by pathogenic organisms. Microbes have the ability to circumvent the mechanism of antibiotic drugs resulting in the development of antimicrobial resistance. Many available evidences showed that the frequent use of large amounts of conventional antibiotics result in drug resistant pathogens, particularly antibiotic-resistant bacteria. The growing burden of drug-resistant infections demand for suitable antimicrobial/antibiotic alternatives. AMPs are ubiquitous short peptides that exhibit broad spectrum of potent antimicrobial efficacy and are capable of being applied to treat various microbial infections including drug-resistant ones. AMPs, also referred as peptide antibiotics have gained significant prominence as innovative antibiotics with great importance in the last few years. Several AMPs with potent antimicrobial properties against bacteria, fungi and virus have been documented. These peptides are shown to have antimicrobial activities by obstructing the functionality of biological molecules present in the cell membrane, making the microbes susceptible [1,2]. Hence, instead of conventional antibiotics, these peptides have promising opportunity to develop into antimicrobial drugs. The importance and therapeutic potential of AMPs have been reviewed in detail elsewhere [3,4,5].
AMPs are reported to exhibit a broad spectrum of antiviral, antifungal, antiparasitic, immunomodulatory and anti-angiogenic activities [6,7,8,9]. The anionic/cationic charges, amphiphilic and hydrophobic properties of AMPs were determined by their amino acid composition which in turn show selective effects on the microbes. Though most of the AMPs are cationic with substantial hydrophobic residues, anionic AMPs containing mostly acidic amino acids like aspartic acid and glutamic acid also exist [10,11]. AMPs display antimicrobial activity by a unique mode of action via targeted destruction of the bacterial membrane and/or by translocation into the cytoplasm to neutralize intracellular targets [1,12,13]. The direct activity against bacteria involves strong electrostatic interaction of positively charged AMPs with the negatively charged microbial surface [14,15]. Bacterial membranes comprise abundant number of amphiphilic lipids such as phosphatidylglycerol, cardiolipin, and phosphatidylethanolamine in their cytoplasmic membranes. The head groups of anionic phospholipids are negatively charged and bind strongly to cationic AMPs [15,16]. Moreover, teichoic acids and lipopolysaccharides in gram-positive and gram-negative bacteria, respectively, provide electronegative charge to bacteria making them highly attractive targets for AMPs [17,18]. The interaction of AMP with the microbial membrane eventually results in destroying the microbes. In contrast, mammalian cell membranes differ from microbial membranes by having rich amount of zwitterionic phospholipids such as phosphatidylethanolamine, phosphatidylcholine, and sphingomyelin in their cytoplasmic membranes [19]. These phospholipids produce neutral charge on mammalian membranes resulting in low binding efficiency of AMPs and enable protection and selectivity against mammalian cells [15,19]. Further AMPs bind with mammalian membranes via weak hydrophobic interactions and are rich in cholesterol which reduces activity of these peptides [20].
To date, more than 2000 AMPs have been identified, synthetically designed, engineered accounting from all living forms including prokaryotic and eukaryotic organisms [21]. Native AMPs can be classified according to source, activity, structure and amino acid residues wherein examples include but are not limited to defensins [22], cathelicidins [23], cecropins [24], lactoferricin [25], dermcidin [26], and anionic peptides [11]. Since their discovery, AMPs have been of great scientific interest due to their importance in human health, as natural antibiotic agents, but also due to their potential as innate immune modulators [27].
Due to its therapeutic potential against drug-resistant pathogens, AMPs are considered as next generation of antimicrobials having potential for pharmacological applications. Furthermore, certain AMPs exert a broad spectrum of activity against diseases in plant species caused by different plant pathogens including bacterial, fungal and viral pathogens [28]. Hence, they represent an innovative crop plant protection method for engineering insect or disease resistance traits for sustainable agriculture. Although AMPs are widely found in their natural form, their synthesis involves cumbersome processes that result in low yields and has so far not proven cost-effective [29,30,31,32,33]. Several features of AMP manufacturing limit the commercial development of these peptides. Chemical peptide synthesis based on solid-phase techniques is a mature technology [34] allowing the production of naturally existing or synthetic polypeptides of small size. In the context of green sustainable chemistry, to avoid the use of large amounts of organic solvents in the chemical synthesis of AMPs, water-based solid phase peptide synthesis using water to replace organic solvents has been developed, facilitated by the conversion of amino acids into water-soluble forms. However there exist a few limitations such as the requirement of additional conversion steps, high preparation costs, need of more energy and resources [35,36]. Moreover, this method also faces issue in the production of large peptides with complex post-translational modifications. Contemporary research utilizes a sophisticated approach to produce AMPs. The recombinant production of AMP in heterologous expression systems provide an opportunity for large scale production of AMPs thereby increasing their accessibility and broadening of their applications in the pharmaceutical sector. Genetic engineering strategies have been employed for production of AMPs in microbial and eukaryotic systems. Traditionally bacteria, yeast and mammalian cells were commonly used for recombinant protein production, however plant-expression systems have considerable advantages like flexibility, scalability and speed. Hence in recent decade, plants have become considered an affordable recombinant protein expression platform. Although plants have been widely utilized for producing vaccine antigens, diagnostic reagents and other high-value biopharmaceuticals in recent times, the recombinant production of AMPs in plants are very limited. Therefore, we focus in this review on the importance of AMPs, possibilities and biotechnological approaches for the expression and production of pharmaceutically important AMPs in plants and further discuss the recent progress which has been made in this direction. Overall, some of the major advances in the field with the view to provide insights on the production of AMPs in plants are presented.

2. AMPs for Infection and Therapy

AMPs differ from antibiotics by having broad range and rapid inhibitory activities against bacteria, fungi, parasites, viruses and in their capacity to overcome resistance in microbial populations [18,37]. Most AMPs directly target the bacterial cell membrane which results in comparable levels of action on multidrug-resistant organisms. Consequently, combined treatment with other intracellular drugs postulates no overlap in modes of action and cross-resistance effects. Thus, considering desirable potency and bactericidal ability, AMPs constitute a promising class of therapeutics for the treatment of drug-resistant infections [3].
To date, a number of AMPs are either in pre-clinical and clinical development or approved for commercial applications. Selected AMPs are listed in Table 1. Polymyxins are one of the most well-studied cyclic peptides utilized as clinically available drugs or treatment for eye infections and multidrug resistant pathogens and are also used for selective digestive tract decontamination and local skin infections [38,39]. Gramicidins are another type of cyclic anti-infective peptides used to treat wounds and local infections in the nose, eyes, and throat [40,41]. Daptomycin is one of the cyclic AMP-based drugs recently approved by the FDA for the treatment of complicated skin and skin-structure infections (cSSSI) caused by Staphylococcus aureus [42,43]. Meanwhile, some of the AMPs under clinical trials, including well-characterized peptides—pexiganan and omiganan—are presently being investigated for the treatment of many bacterial and fungal infections. Accordingly, pexiganan is a Xenopus magainin analog targeting bacteria, fungi, and antibiotic-resistant microbes. It has been assessed in phase III clinical trials and administered as a topical cream for diabetic foot ulcers [44] and further examined for cSSSI [42]. Of note, omiganan is a bovine indolicidin analog that has been evaluated in clinical trials and administered as a topical gel for catheter infections, rosacea, dermatitis, genital warts and inflammatory acne vulgaris [3,45,46]. On the contrary, a few AMPs under clinical trials such as LL-37 and PXL-01 are being evaluated for their mode of action unrelated to microbial infections. For LL-37, pre-clinical results showed its role in wound healing in mice [47,48] and wound infections in pigs [49]. In line with these reports, a phase I/II clinical trial using topical treatment with LL-37 markedly promoted wound healing rates in patients with hard-to-heal chronic leg ulcers [50] and has been recommended to regulate re-epithelialization, angiogenesis and inflammatory response [51]. For PXL01, nonclinical evidence revealed anti-adhesion properties [52] and it effectively prevented adhesion formation linked to post-abdominal surgery in rats [53]. A phase II clinical study further established the efficacy of using PXL01 in sodium hyaluronate for inhibiting post-surgical adhesions and improving post-surgical recovery of the hand [54].
Consideration of AMPs for their clinical applications requires that the underlying issues of activity, toxicity and stability be addressed to achieve progress and commercial success. An ideal AMP should display high antimicrobial activity and specificity, less toxicity towards mammalian cells, high stability and low production costs. Despite showing antimicrobial potency against the microbes, in long term use AMPs have undesirable activities by eliciting immediate immunogenic responses, systemic toxicity, hemolytic activity and other side effects in mammalian cells or in vivo animal models. Hence detailed studies are essential to assess the feasibility and safety profile of AMPs before progressing them towards practical application. The therapeutic index, which is calculated as the ratio of the hemolytic activity and antimicrobial activity of AMPs, is a widely used parameter to evaluate the specificity of AMPs against prokaryotic and eukaryotic cells. Thus, higher values of therapeutic index represent greater specificity [55]. Some AMPs have demonstrated nephrotoxicity and neurotoxicity effects [56], frequently associated with high dosages. Further poor stability of AMPs is another major limitation that critically affects their oral administration, as peptide antibiotics are characterized with low oral bioavailability owing to enzymatic degradation and poor permeability in the intestinal mucosa. Likewise, systemic administration via intravenous injection restricts applications of AMPs due to rapid degradation or rapid hepatic and liver clearance resulting in significantly reduced or short half-life [57]. Selective drug delivery methods can address some of the limitations hindering its applications viz., topical application of AMPs may reduce the systemic toxicity and proteolytic stability [58]. To overcome the limitations and multiple restrictive factors, many approaches have been employed to develop ideal AMPs, including multi-disciplinary strategies with computational/bioinformatic tools, biophysical experiments and biological validations which are discussed in detail elsewhere [59,60].
Table 1. List of few AMPs at various stages of clinical trials.
Table 1. List of few AMPs at various stages of clinical trials.
Anti-Microbial PeptideIn Vivo/Clinical/ApprovedIndicationAdministrationReference
Mutacin B-Ny266 (lantibiotic)In vivoMulti-drug resistant bacteria infection-[61]
Actagardine (lantibiotic)In vivoStaphylococcal, enterococcal, C. difficile infections-[62]
Plectasin (defensin)In vivoSystemic pneumococcal and streptococcal infections-[63]
Planosporicin (lantibiotic)In vivoStaphylococcal and enterococcal infections -[64]
Gallidermin/Epidermin (lantibiotic)In vivoAcne, eczema, folliculitis, and impetigo-[65]
Microbisporicin (lantibiotic)In vivoStaphylococcal and enterococcal infections; Acne-[66]
Mersacidin (lantibiotic)In vivoStaphylococcal, enterococcal, Clostridioides difficile infections-[67]
Lacticin 3147 (lantibiotic)In vivoStaphylococcal and enterococcal infections; Acne-[68]
Salivaricin B (lantibiotic)In vivoStreptococcal infections (caused mainly by S. pyogenes) and dental caries-[69]
Duramycin (lantibiotic)In vivoCystic fibrosis, ocular diseases, and disorders-[70]
Deoxyactagardine/NVB302 (lantibiotic)In vivoC. difficile infections-[71]
Nisin (lantibiotic)In vivoStaphylococcal and enterococcal infections-[72]
Pinensins (lantibiotic)In vivoYeast/fungal infections-[73]
MX-226In vivoCatheter infections-[74]
PAC-113 (histatin 3)Phase II
Identifier: NCT00659971
Oral candidiasis in HIV patientsOral (Mouthwash)[75]
Omiganan (indolicidin)Phase III
Identifier:
NCT00231153
Prevent local site catheter infection and colonization with central venous cathetersTopical[46]
Iseganan (protegrin-1)Phase II
Identifier: NCT00118781
Ventilator-associated pneumoniaOral (Mouthwash)[76]
Phase III
Identifier: NCT00022373
Oral mucositis induced by chemotherapyOral (Mouthwash)[77]
Pexiganan (magainin analog)Phase III
Identifier: NCT00563-394/433
Diabetic foot ulcer infectionsTopical[44]
hLF1-11 (lactoferrin)Phase I/II
Identifier: NCT00509938
Bacteraemia and fungal infectionIntravenous[78]
CZEN-002 (α-melanocyte-stimulating hormone)Phase IIbVaginal candidiasisVaginal gel[79]
Novexatin (defensin)Phase II
Identifier: NCT02343627
Stubborn fungal nail infectionTopical[45]
LL-37 (cathelicidin)Phase I/II
Identifier: NCT04098562
Hard-to-heal venous leg ulcersTopical[50]
PXL01 (lactoferricin)Phase II
Identifier: NCT01022242
Prevent post-operative adhesion in handsHydrogel applied at surgical site[54]
Surotomycin (synthetically modified daptomycin)Phase III
Identifier: NCT01597505
Diarrhea caused by C. difficileOral[80]
LTX-109 (synthetic antimicrobial peptidomimetic)Phase II
Identifier: NCT01803035
Skin infection, impetigo Topical[81]
Phase I/II
Identifier: NCT01158235
Nasal infection with S. aureusNasal[81]
SGX942 (indolicidin)Phase III
Identifier: NCT03237325
Oral mucositis induced by radiation and/or chemotherapyIntravenous[82]
OP-145 (cathelicidin)Phase I/IIChronic otic infectionEardrops[83]
C16G2 (synthetic specific-directed antimicrobial peptide)Phase II
Identifier: NCT02044081
Avoid caries caused by S. mutansOral (Mouthwash)[84]
Murepavadin (protegrin I)Phase I
Identifier: NCT03409679
Ventilator-associated pneumonia and bronchiectasis
by Pseudomonas aeruginosa
Intravenous[85]
DPK-060 (hybrid peptide from 2 functional domains)Phase II
Identifier: NCT01522391
Human wound infection caused by S. aureusTopical[86]
Teicoplanin (Actinoplanes teichomyceticus glycopeptide)ApprovedBacterial infectionsIntravenous and Intramuscular[87]
Daptomycin (anionic peptide)ApprovedBacterial skin infectionsIntravenous[43]
Colistin (Bacillus polymyxa cyclic peptide)ApprovedMulti drug-resistant gram-negative infectionsIntravenous[88]
Dalbavancin (Teicoplanin derivative lipoglycopeptide)ApprovedAcute bacterial skin and skin structure infectionsIntravenous[89]
Polymyxin (Bacillus polymyxa polypeptide)ApprovedUrinary tract and bloodstream infectionsOphthalmic Topical Intravenous[38]
Enfuvirtide (biomimetic peptide)ApprovedHIV-1 infectionSubcutaneous[90]
Telavancin (vancomycin derivative lipoglycopeptide)ApprovedBacterial skin infectionsIntravenous[91]
Gramicidin D (Bacillus brevis polypeptides)ApprovedSkin and eye infectionsTopical Ophthalmic[40]
Oritavancin (vancomycin derivative lipoglycopeptide)ApprovedBacterial skin infectionsIntravenous[92]
Bacitracin (Bacillus licheniformis cyclic peptide)ApprovedSkin and eye infections; wound infectionsTopical[93]
Telaprevir (antimicrobial peptidomimetic)ApprovedHepatitis C infectionOral[94]
Vancomycin (Amycolatopsis orientalis glycopeptide)ApprovedBacterial infectionsOral and Intravenous[95]

3. Heterologous Production of AMPs

Recent advances in recombinant DNA engineering provide an insight for the economical production of AMPs in various heterologous host systems [96,97,98,99]. Furthermore, recombinant expression platforms will undoubtedly speed up the approaches for developing novel peptide therapeutics and are also helpful for the betterment of existing ones. Many expression hosts are currently available for the production of various short AMPs with varied sizes, folds and complexities. Certain factors like size, intracellular localization, secretion, protein folding and glycosylation need to be considered during the selection of a host expression system to produce AMPs. Microbial systems (bacteria and yeast) are the most widely employed as they are easy to manipulate and have rapid growth rates, multiplication times and high cell densities [100]. Bacterial species such as Escherichia coli, Bacillus subtilis, Propionibacterium freudenreichii were used for expression of different AMPs like adenoregulin, cecropin, crustin, defensin, hepcidin, histonin, human β defensin, lactoferrin, perinerin, thanatin and viscotoxin [101]. E. coli was the most prominent bacterium used to express AMPs, due to its easy growth rate, well developed recombinant methods for its manipulation and the abundant available literature on its genetic morphology and physiology [2,63]. Though many AMPs are expressed in bacteria, there are few hurdles that need to be addressed in order to achieve efficient production. The produced AMPs that have natural activity must be prevented from exerting their lethal action on the host strain. A lack of post-translational modification and the need for carrier/fusion proteins are other issues [2,63,102,103]. Pichia pastoris and Saccharomyces cerevisiae are commonly used for the expression of AMPs such as the antifungal proteins cathelicidin, enterocin, pediocin, plantaricin, and α-sarcin [101,104,105]. Nevertheless, plant systems have also been utilized for AMP production in recent decades. Tobacco is one of the most highly explored leaf-based production platforms for recombinant protein expression.

4. Plant Molecular Farming

The expression of AMPs in plants presents a dual role as their antimicrobial activity helps in plant protection while also meeting the demand for novel antimicrobial agents in the biopharmaceutical industry [106]. The process of utilizing plants and plant cell cultures as an effective production platform for recombinant proteins with industrial or pharmaceutical significance is called molecular farming and the protein products are often referred to as plant-made pharmaceuticals (PMPs). Plants act as remarkable hosts for producing various recombinant proteins due to their many advantages over other prokaryotic and eukaryotic expression systems. The major propitious features include the cheaper cost, high yields with the feasibility for easy scale up, simple manufacturing methods, minimizing the extensive purification and processing techniques in the case of oral vaccines [107,108,109]. The plant-made vaccines or therapeutic products can be easily stored or lyophilized for longer shelf life without the requirement of low temperatures for keeping them stable and retaining their activity [110]. Further, the pharmaceutically-relevant proteins produced in plants are considered to be safer when compared to bacterial or mammalian cells as the risk of contamination during the manufacturing processes is low and the do not present serious bio-safety threats [111]. The major advantage of this expression system is the ability to perform post-translational modifications which are likely crucial for protein folding and the biological function of AMP molecules [112,113]. The advantages and challenges of different protein production systems are summarized in Table 2.
In 1986, the recombinant human growth hormone was the first plant-derived pharmaceutically-relevant protein produced in transgenic tobacco and sunflower [115], followed by the report of functional antibody expression in transgenic tobacco plants [116]. After two decades of research, the first PMP “Elelyso” (recombinant β-glucocerebrosidase) produced in carrot suspension culture was approved by FDA in 2012 for the treatment of Gaucher’s disease. In addition, the regulatory approval of tobacco-produced HIV-neutralizing human monoclonal antibody 2G12 established the scientific, technical and regulatory framework for plant-derived recombinant proteins [117]. Recently, virus-like particle (VLP) influenza vaccine manufactured in plants has completed the phase III trial and plant-derived VLP vaccine for coronavirus disease produced in N. benthamiana has completed a Phase I trial [118]. Furthermore, number of PMPs are in various stages of clinical development which include vaccine antigens, enzymes, cytokines, monoclonal antibodies and their fragments and a few are approved [119,120]. The approval of the first PMP in the commercial market and promising results of plant-proteins in clinical trials pave the way for the further development of recombinant products.
Several examples of such PMPs and the detailed advantages, limitations and challenges of plant expression system for the production of the desired targets have been comprehensively reviewed elsewhere [121,122,123,124,125,126,127,128]. The different plant-based expression systems range from transgenic plants to cell suspensions cultures are available for AMP production in plants [129,130,131,132,133] which are described briefly in the following sections.

5. Strategies for Protein Production in Plants

The production of recombinant proteins in plants form an ideal cost-effective platform gaining attraction for commercial biopharmaceutical production. The strategies employed for recombinant protein production in plants are stable expression, transient expression and suspension cell cultures (Figure 1). The stable expression is a conventional method of recombinant technology which involves the incorporation of foreign genes into nucleus for nuclear genomic expression [134] and/or to chloroplast for plastid genomic expression [135] resulting in the generation of stable transgenic/transplastomic plant lines.

5.1. Stable Nuclear Expression

Nuclear expression facilitates the stable integration of transgenes into the nuclear genome of plant cells. This technique is regulated by transcription of the gene of interest in the nucleus and then translation in the cytoplasm [136]. The most widely used gene delivery system into the plant is via Agrobacterium tumefaciens-mediated transformation. The plant bacterium, A. tumefaciens, has the capability to deliver a particular DNA segment (T-DNA) into the plant nucleus which is commonly localized on the tumor-inducing (Ti) plasmid of Agrobacterium [137]. Meanwhile, plant transformation utilizes a binary vector system devised according to the T-DNA of Agrobacterium. Consequently, the T-DNA-containing gene expression cassette is separated from the vector backbone and transformed into plants, permitting easy genetic engineering of plants. This vector system is introduced into a modified Agrobacterium (not containing bacterial genes within the T-DNA region) to infect the plant cells or tissues and transfer the T-DNA-containing gene of interest from the binary vector for expression in nuclear genome of plant host [138]. These tissues are cultivated in an antibiotic-containing growth medium, allowing selective growth of transformants harboring the gene of interest. Then, growth of callus tissue and development of shoots and roots are observed. Upon successful rooting, plantlets are transferred to the soil and the expression of foreign genes in the transgenic lines can be characterized [139]. The Agrobacterium-mediated transformation offers the simplest and a conventional method for genetic modification of crops with the horizontal transgene transfer and consistent recombinant protein expression [140]. However, few associated disadvantages of this method includes gene silencing, transgene contamination risk, potential interactions with natural products, low yields (about <1% of total soluble protein) and time-consuming genetic manipulation [136,141,142]. An early study of pharmaceutical recombinant antibodies stably integrated and produced in transgenic tobacco plants was recorded in 1989. Since then, several proteins have been produced in stably transformed plants, including anti-cancer agents [143,144] and antimicrobial peptides as anti-infectives [145]. AMPs with anti-bacterial or anti-fungal properties can be stably expressed in plants which confers disease resistance against plant pathogens which in turn increases the yield, quality and safety of agricultural products.

5.2. Stable Chloroplast Expression

Chloroplast expression directs incorporation of transgenes into the chloroplast genome of plant cells. This approach effectively transforms foreign genes into plant chloroplasts by using a particle gun or gene gun or biolistic transformation method. In particular, plant tissues are bombarded with DNA-coated gold or tungsten particles [146]. Then, plant tissues are cultivated in suitable growth medium supplemented with appropriate antibiotics, which confer selection of transformants containing the gene of interest. Similar like nuclear transformation, characterization of callus formation, shoots and roots development is observed. The young plantlets are transplanted into soil to generate mature transplastomic plants. Chloroplast transformation of recombinant gene offers several advantages compared to nuclear transformation. The chloroplast genome provides ease of manipulation as DNA-containing cassette can be inserted in between functional chloroplast genes by homologous recombination [147]. The specific targeting ensures high levels of expression and prevents gene placement into a poorly transcribed region of the genome. Due to high copies of chloroplast in plant cells, optimal yields of recombinant proteins have been attained by chloroplast expression [136,147]. Additional advantages include no risk of transgene contamination or leakage into the environment since chloroplast genes are maternally inherited [146], a neutral pH and low number of active proteases [148]. The expression of therapeutic proteins in chloroplast has been well-explored for several antigens, growth factors, interferons and many other pharmaceutical proteins [140,149,150,151].

5.3. Transient Expression

Transient gene expression involves rapid production of recombinant proteins without chromosomal integration into the plant cell genome. It has been employed as an approach for determining the expression efficiency of transgenes in the plant nucleus after a short period of time [152]. There are two promising methods of transient expression involving plant pathogen vectors, namely, plant viruses (plant viral infection) and A. tumefaciens (agroinfiltration). Plant virus-mediated transient expression directs amplification of viral vectors within plant cells and introduces the transgene of interest by utilizing mature viral particles [153]. Examples of plant virus-based vectors include but not restricted to tobamoviruses, potexviruses, potyviruses, bromoviruses, comoviruses, and geminiviruses [154,155,156]. This strategy has shown advantages and suitability to produce several plant-based biopharmaceuticals [157]. In addition, they have been characterized for their fast transmission from one plant cell to another, yielding high expression efficiency [158]. Meanwhile, risk of viral vector contamination in plants and the environment has to be regarded with careful consideration [159].
Agrobacterium-mediated transient expression refers to the process of infecting plant leaves with Agrobacterium cells suspension containing T-DNAs with gene of interest [160]. It can be performed either by infiltration using syringe without a needle (syringe infiltration) or by large-scale infiltration (vacuum infiltration). Agrobacterium infection spreads across the site of injection and infected plants can be harvested within few days of post-infiltration. Transient approach is the method of choice for the scalable production of AMPs for large scale applications in the food industry (as preservative), as topical disinfectant or as a feed supplement for livestock or poultry [161].

5.4. Suspension Cultures

Suspension plant cell cultures are more promising and ideal platforms than using the whole plants to produce various important biological active products. These cultures are grown in controlled environments under monitoring and defined conditions for the growth of plant cells thus complying all the regulatory concern. The plant cells are cultured in aseptic in vitro growth conditions in a sterile sealed container without any human or microbial contaminants [162,163]. Other biosafety and environmental issues can also be overcome by using plant bioreactors preventing cross fertilization and transmission of pollen. The production costs for recombinant proteins using plant suspension cell cultures are quite low in comparison to mammalian and bacterial systems as they require simple growth media and nutritional requirements [164]. The downstream purification and processing of plant produced products does not require any complex methods [165]. The first FDA-approved plant-produced pharmaceutical taliglucerase-α was produced in carrot suspension cell cultures almost reducing the conventional orphan drug treatment costs by 75% [166]. Other popular plant cells include BY-2 and NT-1 tobacco strains that are used as bioreactors where the proteins can be secreted into the culture medium simplifying the purification process. BY-2 cell cultures were used to produce human monoclonal antibody M12 in a 200 L bioreactor yielding 20 mg/L of the mAb [164,167]. The BY-2 cell lines have the capacity to multiply up to 100-fold in one week with a generation time of 16–24 h under defined growth parameters [168]. Suspension cultures hold significant potential in therapeutic AMP production for medical applications as they are easy to scale-up, compliant with GMP and they meet regulatory requirements for biopharmaceutical production.

6. AMP Expression in Plants

Several AMPs have been expressed in plants with the perspective of clinical and agricultural development. Plants have come into limelight for the expression of AMPs in desired crop plants for direct defense against pathogens and also large-scale and cost-effective production of recombinant AMPs. Although protein accumulation varies between the AMP expressed, but the functional activity of the recombinant AMP confirmed its active form. Protegrin-1, a broad-spectrum AMP was expressed in low alkaloid tobacco species using transient approach and was found to be effective against K. pneumoniae, S. aureus, E. coli, M. bovis BCG, and C. albicans [169]. Lfchimera, a chimerical peptide was codon optimized, expressed in plant culture system and tobacco hairy roots in vitro and significant antimicrobial activity was reported against clinical and phytopathogenic bacteria [170,171]. AMPs apart from exhibiting antimicrobial activity, they were further investigated for inducing resistance against various bacterial and fungal pathogens in plants [162]. The peptide LL-37 was produced in transgenic barley by expressing codon optimized chimeric LL-37 under the influence of endosperm specific promoter of barley B1 hordein gene, accumulating upto 0.55 mg/kg of grain and the plant-produced LL-37 was biologically active [172]. An insect antimicrobial peptide, thanatin S, was expressed by fusing with signal peptide of rice Cht1 in Arabidopsis, that showed enhanced resistance to phytopathogenic fungi and bacteria [173]. In a study by Jung et al., human cathelicidin hCAP18 was expressed in Chinese cabbage fusing the DNA encoding fragment for this AMP with C-terminal end of endopolygalacturonase inhibiting protein under the control of CaMV 35 S promoter showing varied levels of resistance to bacterial and fungal pathogens [174]. Two proteins, snakin-2 (SN2) a cysteine-rich peptide and extensin-like protein (ELP) a major cell-wall hydroxyproline-rich glycoprotein were over expressed in tomato cultivars and showed resistance against Clavibacter michiganensis subsp. michiganensis (Cmm) [175]. A snakin-1 gene isolated from potato was found to have in vitro antimicrobial activity and when transformed into wheat by particle bombardment, showed effective protection against soil borne fungus Gaeumannomyces graminis var. tritici which causes root disease [176]. Defensins, SmAMP 2 gene, sarcotoxin IA, retrocyclin 101, hevein like peptides, C4V3, trichokonins, cecropin B, temporin A, snakin-2, cathelicidins and MsrA2 were also expressed in different plant species as listed in Table 3.

7. Conclusions

Diverse AMPs hold major potential for the development of innovative approaches in both clinical and agricultural biotechnology. Disease-resistant plant traits developed by introducing AMPs might increase yields and offer safety of agricultural products against phytopathogens. Further recombinant expression of AMPs in plant platforms overcome the limitations associated with the large-scale production of these recombinant peptides for clinical use. The urgent need of rapid, cost-effective protein production systems for the production of large amounts of recombinant protein has been driving the plant molecular farming research. There has been much progress in our understanding of this field and extensive research has been performed over the last three decades on plant-based biopharmaceutical production against various pathogens. The plant-derived proteins are shown to be functional and even shown to be effective in clinical trials. However, plant-made pharmaceuticals still encounter some technological and regulatory issues limiting prospective investors eventually resulting in a long timeframe of potential products from bench-to-market. Despite the many proof-of-concept studies, few products are approved for commercial applications. The challenges faced by PMPs during initial stages of plant molecular farming such as longer production time, transgene escape and safety have been addressed in recent decades. The issues related to low yield and time associated with the stable expression have been addressed by developing transient expression systems. Thus, the proper selection of expression strategy, vector, and extraction/purification techniques is essential to achieve high product yield, desired functionality, safety and quality of the products. Furthermore, the recent advances in the plant biotechnology have pushed various regulatory bodies to develop regulatory frameworks for the process of genetic transformations or on the final plant-derived product [208]. There is substantial evidence showing the capability of making proteins with high quality to address a range of human health-related issues particularly in low-income and middle-income countries. The FDA approval for the therapeutic enzyme Elelyso was a major milestone in the field. Most likely, we can expect a significant number of plant-derived biopharmaceuticals on the market in the upcoming years. There are several promising AMPs which are in different stages of clinical trials. These AMPs could be potential candidates for plant-based manufacturing. Thus, the integration of our existing knowledge of the plant biotechnology, huge strides that have been made in plant transient expression and glycoengineering strategies coupled with the design, development and accessibility of AMPs could make an ideal foundation for the design of a novel class of plant-derived AMP based therapeutics that hold promising potential. In summary, biotechnological perspectives for the rapid large-scale production of AMPs in plant systems has been provided. The existing knowledge on plant expression system opens the way to produce and evaluate the potentiality of AMPs that could be rapidly manufactured, at low cost and with negligible risk, to fight against drug resistant pathogens in post-antibiotic era. Altogether production of AMPs in plants is considered a prospective tool for novel applications in medicine and agriculture.

Author Contributions

Conceptualization, B.S. and W.P.; literature review, B.S., C.J.I.B., A.M.; writing—original draft preparation, B.S., C.J.I.B., A.M.; writing—review and editing, B.S., C.J.I.B., A.M.; supervision, W.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

C.J.I.B. was supported by the Scholarship Program for ASEAN and Non-ASEAN Countries, Chulalongkorn University.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Hancock, R.E.W.; Sahl, H.-G. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat. Biotechnol. 2006, 24, 1551–1557. [Google Scholar] [CrossRef]
  2. Li, Y. Recombinant production of antimicrobial peptides in Escherichia coli: A review. Protein Expr. Purif. 2011, 80, 260–267. [Google Scholar] [CrossRef]
  3. Mahlapuu, M.; Björn, C.; Ekblom, J. Antimicrobial peptides as therapeutic agents: Opportunities and challenges. Crit. Rev. Biotechnol. 2020, 40, 978–992. [Google Scholar] [CrossRef]
  4. Pfalzgraff, A.; Brandenburg, K.; Weindl, G. Antimicrobial peptides and their therapeutic potential for bacterial skin infections and wounds. Front. Pharmacol. 2018, 9, 281. [Google Scholar] [CrossRef] [PubMed]
  5. Lei, J.; Sun, L.; Huang, S.; Zhu, C.; Li, P.; He, J.; Mackey, V.; Coy, D.H.; He, Q. The antimicrobial peptides and their potential clinical applications. Am. J. Transl. Res. 2019, 11, 3919–3931. [Google Scholar] [PubMed]
  6. Hoskin, D.W.; Ramamoorthy, A. Studies on anticancer activities of antimicrobial peptides. Biochim. Biophys. Acta BBA Biomembr. 2008, 1778, 357–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Li, C.; Blencke, H.-M.; Paulsen, V.; Haug, T.; Stensvåg, K. Powerful workhorses for antimicrobial peptide expression and characterization. Bioeng. Bugs 2010, 1, 217–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Silva, O.N.; Mulder, K.C.; Barbosa, A.E.; Otero-Gonzalez, A.J.; Lopez-Abarrategui, C.; Rezende, T.M.; Dias, S.C.; Franco, O.L. Exploring the pharmacological potential of promiscuous host-defense peptides: From natural screenings to biotechnological applications. Front. Microbiol. 2011, 2, 232. [Google Scholar] [CrossRef] [Green Version]
  9. Thevissen, K.; Kristensen, H.H.; Thomma, B.P.; Cammue, B.P.; François, I.E. Therapeutic potential of antifungal plant and insect defensins. Drug Discov. Today 2007, 12, 966–971. [Google Scholar] [CrossRef]
  10. Lai, Y.; Villaruz, A.E.; Li, M.; Cha, D.J.; Sturdevant, D.E.; Otto, M. The human anionic antimicrobial peptide dermcidin induces proteolytic defence mechanisms in staphylococci. Mol. Microbiol. 2007, 63, 497–506. [Google Scholar] [CrossRef]
  11. Harris, F.; Dennison, S.R.; Phoenix, D.A. Anionic antimicrobial peptides from eukaryotic organisms. Curr. Protein Pept. Sci. 2009, 10, 585–606. [Google Scholar] [CrossRef]
  12. Martin, M. Interactions of antimicrobial peptides with bacterial membranes and membrane components. Curr. Top. Med. Chem. 2016, 16, 16–24. [Google Scholar] [CrossRef]
  13. Wang, J.; Dou, X.; Song, J.; Lyu, Y.; Zhu, X.; Xu, L.; Li, W.; Shan, A. Antimicrobial peptides: Promising alternatives in the post feeding antibiotic era. Med. Res. Rev. 2019, 39, 831–859. [Google Scholar] [CrossRef]
  14. Yeung, A.T.; Gellatly, S.L.; Hancock, R.E. Multifunctional cationic host defence peptides and their clinical applications. Cell. Mol. Life Sci. CMLS 2011, 68, 2161–2176. [Google Scholar] [CrossRef]
  15. Ebenhan, T.; Gheysens, O.; Kruger, H.G.; Zeevaart, J.R.; Sathekge, M.M. Antimicrobial peptides: Their role as infection-selective tracers for molecular imaging. BioMed Res. Int. 2014, 2014, 867381. [Google Scholar] [CrossRef] [PubMed]
  16. Lin, T.Y.; Weibel, D.B. Organization and function of anionic phospholipids in bacteria. Appl. Microbiol. Biotechnol. 2016, 100, 4255–4267. [Google Scholar] [CrossRef]
  17. Bechinger, B.; Gorr, S.U. Antimicrobial peptides: Mechanisms of action and resistance. J. Dent. Res. 2016, 96, 254–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Bahar, A.A.; Ren, D. Antimicrobial peptides. Pharmaceuticals 2013, 6, 1543–1575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Yeaman, M.R.; Yount, N.Y. Mechanisms of antimicrobial peptide action and resistance. Pharmacol. Rev. 2003, 55, 27–55. [Google Scholar] [CrossRef] [Green Version]
  20. Zasloff, M. Antimicrobial peptides of multicellular organisms. Nature 2002, 415, 389–395. [Google Scholar] [CrossRef] [PubMed]
  21. Waghu, F.H.; Idicula-Thomas, S. Collection of antimicrobial peptides database and its derivatives: Applications and beyond. Protein Sci. 2020, 29, 36–42. [Google Scholar] [CrossRef]
  22. Reddy, K.V.R.; Yedery, R.D.; Aranha, C. Antimicrobial peptides: Premises and promises. Int. J. Antimicrob. Agents 2004, 24, 536–547. [Google Scholar] [CrossRef]
  23. D’Este, F.; Benincasa, M.; Cannone, G.; Furlan, M.; Scarsini, M.; Volpatti, D.; Gennaro, R.; Tossi, A.; Skerlavaj, B.; Scocchi, M. Antimicrobial and host cell-directed activities of Gly/Ser-rich peptides from salmonid cathelicidins. Fish Shellfish Immunol. 2016, 59, 456–468. [Google Scholar] [CrossRef] [PubMed]
  24. Dutta, P.; Sahu, R.K.; Dey, T.; Lahkar, M.D.; Manna, P.; Kalita, J. Beneficial role of insect-derived bioactive components against inflammation and its associated complications (colitis and arthritis) and cancer. Chem. Biol. Interact. 2019, 313, 108824. [Google Scholar] [CrossRef] [PubMed]
  25. Sibel Akalın, A. Dairy-derived antimicrobial peptides: Action mechanisms, pharmaceutical uses and production proposals. Trends Food Sci. Technol. 2014, 36, 79–95. [Google Scholar] [CrossRef]
  26. Zeth, K.; Sancho-Vaello, E. The human antimicrobial peptides dermcidin and LL-37 show novel distinct pathways in membrane interactions. Front. Chem. 2017, 5, 86. [Google Scholar] [CrossRef]
  27. Kosikowska, P.; Lesner, A. Antimicrobial peptides (AMPs) as drug candidates: A patent review (2003–2015). Expert Opin. Ther. Pat. 2016, 26, 689–702. [Google Scholar] [CrossRef]
  28. Rahnamaeian, M.; Langen, G.; Imani, J.; Khalifa, W.; Altincicek, B.; von Wettstein, D.; Kogel, K.-H.; Vilcinskas, A. Insect peptide metchnikowin confers on barley a selective capacity for resistance to fungal ascomycetes pathogens. J. Exp. Bot. 2009, 60, 4105–4114. [Google Scholar] [CrossRef] [Green Version]
  29. Laverty, G.; Gorman, S.P.; Gilmore, B.F. The potential of antimicrobial peptides as biocides. Int. J. Mol. Sci. 2011, 12, 6566–6596. [Google Scholar] [CrossRef] [Green Version]
  30. Maira Galdino da Rocha, P.; Marina Galdino da Rocha, P.; Suely Lins, G. Development of novel therapeutic drugs in humans from plant antimicrobial peptides. Curr. Protein Pept. Sci. 2010, 11, 236–247. [Google Scholar] [CrossRef]
  31. Peters, B.M.; Shirtliff, M.E.; Jabra-Rizk, M.A. Antimicrobial peptides: Primeval molecules or future drugs? PLoS Pathog. 2010, 6, e1001067. [Google Scholar] [CrossRef] [PubMed]
  32. Li, C.; Haug, T.; Styrvold, O.B.; Jørgensen, T.; Stensvåg, K. Strongylocins, novel antimicrobial peptides from the green sea urchin, Strongylocentrotus droebachiensis. Dev. Comp. Immunol. 2008, 32, 1430–1440. [Google Scholar] [CrossRef] [PubMed]
  33. Barashkova, A.S.; Rogozhin, E.A. Isolation of antimicrobial peptides from different plant sources: Does a general extraction method exist? Plant Methods 2020, 16, 143. [Google Scholar] [CrossRef]
  34. Amblard, M.; Fehrentz, J.-A.; Martinez, J.; Subra, G. Methods and protocols of modern solid phase peptide synthesis. Mol. Biotechnol. 2006, 33, 239–254. [Google Scholar] [CrossRef]
  35. Hojo, K.; Hara, A.; Kitai, H.; Onishi, M.; Ichikawa, H.; Fukumori, Y.; Kawasaki, K. Development of a method for environmentally friendly chemical peptide synthesis in water using water-dispersible amino acid nanoparticles. Chem. Cent. J. 2011, 5, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Martin, V.; Egelund, P.; Johansson, H.; Quement, S.; Wojcik, F.; Pedersen, D. Greening the synthesis of peptide therapeutics: An industrial perspective. RSC Adv. 2020, 10, 42457–42492. [Google Scholar] [CrossRef]
  37. Schweizer, F. Cationic amphiphilic peptides with cancer-selective toxicity. Eur. J. Pharmacol. 2009, 625, 190–194. [Google Scholar] [CrossRef]
  38. Zavascki, A.P.; Goldani, L.Z.; Li, J.; Nation, R.L. Polymyxin B for the treatment of multidrug-resistant pathogens: A critical review. J. Antimicrob. Chemother. 2007, 60, 1206–1215. [Google Scholar] [CrossRef] [Green Version]
  39. Rigatto, M.H.; Falci, D.R.; Zavascki, A.P. Clinical use of polymyxin B. Adv. Exp. Med. Biol. 2019, 1145, 197–218. [Google Scholar] [CrossRef]
  40. Burkhart, B.M.; Gassman, R.M.; Langs, D.A.; Pangborn, W.A.; Duax, W.L.; Pletnev, V. Gramicidin D conformation, dynamics and membrane ion transport. Biopolymers 1999, 51, 129–144. [Google Scholar] [CrossRef]
  41. Cynthia, L.S. Advances in peptide pharmaceuticals. Curr. Pharm. Biotechnol. 2009, 10, 122–137. [Google Scholar] [CrossRef]
  42. Sierra, J.M.; Fusté, E.; Rabanal, F.; Vinuesa, T.; Viñas, M. An overview of antimicrobial peptides and the latest advances in their development. Expert Opin. Biol. Ther. 2017, 17, 663–676. [Google Scholar] [CrossRef] [PubMed]
  43. Tedesco, K.L.; Rybak, M.J. Daptomycin. Pharmacotherapy 2004, 24, 41–57. [Google Scholar] [CrossRef]
  44. Lipsky, B.A.; Holroyd, K.J.; Zasloff, M. Topical versus systemic antimicrobial therapy for treating mildly infected diabetic foot ulcers: A randomized, controlled, double-blinded, multicenter trial of pexiganan cream. Clin. Infect. Dis. 2008, 47, 1537–1545. [Google Scholar] [CrossRef] [Green Version]
  45. Fox, J.L. Antimicrobial peptides stage a comeback. Nat. Biotechnol. 2013, 31, 379–382. [Google Scholar] [CrossRef]
  46. Sader, H.; Fedler, K.; Rennie, R.; Stevens, S.; Jones, R. Omiganan pentahydrochloride (MBI 226), a topical 12-amino-acid cationic peptide: Spectrum of antimicrobial activity and measurements of bactericidal activity. Antimicrob. Agents Chemother. 2004, 48, 3112–3118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Ramos, R.; Silva, J.P.; Rodrigues, A.C.; Costa, R.; Guardão, L.; Schmitt, F.; Soares, R.; Vilanova, M.; Domingues, L.; Gama, M. Wound healing activity of the human antimicrobial peptide LL37. Peptides 2011, 32, 1469–1476. [Google Scholar] [CrossRef] [Green Version]
  48. Kos, S.; Vanvarenberg, K.; Dolinsek, T.; Cemazar, M.; Jelenc, J.; Préat, V.; Sersa, G.; Vandermeulen, G. Gene electrotransfer into skin using noninvasive multi-electrode array for vaccination and wound healing. Bioelectrochemistry 2017, 114, 33–41. [Google Scholar] [CrossRef]
  49. Steinstraesser, L.; Hirsch, T.; Schulte, M.; Kueckelhaus, M.; Jacobsen, F.; Mersch, E.A.; Stricker, I.; Afacan, N.; Jenssen, H.; Hancock, R.E.; et al. Innate defense regulator peptide 1018 in wound healing and wound infection. PLoS ONE 2012, 7, e39373. [Google Scholar] [CrossRef] [Green Version]
  50. Grönberg, A.; Mahlapuu, M.; Ståhle, M.; Whately-Smith, C.; Rollman, O. Treatment with LL-37 is safe and effective in enhancing healing of hard-to-heal venous leg ulcers: A randomized, placebo-controlled clinical trial. Wound Repair Regen. 2014, 22, 613–621. [Google Scholar] [CrossRef]
  51. Hancock, R.E.W.; Haney, E.F.; Gill, E.E. The immunology of host defence peptides: Beyond antimicrobial activity. Nat. Rev. Immunol. 2016, 16, 321–334. [Google Scholar] [CrossRef] [PubMed]
  52. Edsfeldt, S.; Holm, B.; Mahlapuu, M.; Reno, C.; Hart, D.A.; Wiig, M. PXL01 in sodium hyaluronate results in increased PRG4 expression: A potential mechanism for anti-adhesion. Upsala J. Med. Sci. 2016, 122, 28–34. [Google Scholar] [CrossRef] [PubMed]
  53. Nilsson, E.; Björn, C.; Sjöstrand, V.; Lindgren, K.; Münnich, M.; Mattsby-Baltzer, I.; Ivarsson, M.L.; Olmarker, K.; Mahlapuu, M. A novel polypeptide derived from human lactoferrin in sodium hyaluronate prevents postsurgical adhesion formation in the rat. Ann. Surg. 2009, 250, 1021–1028. [Google Scholar] [CrossRef] [PubMed]
  54. Wiig, M.E.; Dahlin, L.B.; Fridén, J.; Hagberg, L.; Larsen, S.E.; Wiklund, K.; Mahlapuu, M. PXL01 in sodium hyaluronate for improvement of hand recovery after flexor tendon repair surgery: Randomized controlled trial. PLoS ONE 2014, 9, e110735. [Google Scholar] [CrossRef] [Green Version]
  55. Jiang, Z.; Vasil, A.I.; Hale, J.D.; Hancock, R.E.W.; Vasil, M.L.; Hodges, R.S. Effects of net charge and the number of positively charged residues on the biological activity of amphipathic alpha-helical cationic antimicrobial peptides. Biopolymers 2008, 90, 369–383. [Google Scholar] [CrossRef]
  56. Falagas, M.E.; Kasiakou, S.K. Toxicity of polymyxins: A systematic review of the evidence from old and recent studies. Crit. Care 2006, 10, R27. [Google Scholar] [CrossRef] [Green Version]
  57. Vlieghe, P.; Lisowski, V.; Martinez, J.; Khrestchatisky, M. Synthetic therapeutic peptides: Science and market. Drug Discov. Today 2010, 15, 40–56. [Google Scholar] [CrossRef]
  58. Browne, K.; Chakraborty, S.; Chen, R.; Willcox, M.D.; Black, D.S.; Walsh, W.R.; Kumar, N. A new era of antibiotics: The clinical potential of antimicrobial peptides. Int. J. Mol. Sci. 2020, 21, 7047. [Google Scholar] [CrossRef] [PubMed]
  59. Cardoso, P.; Glossop, H.; Meikle, T.G.; Aburto-Medina, A.; Conn, C.E.; Sarojini, V.; Valery, C. Molecular engineering of antimicrobial peptides: Microbial targets, peptide motifs and translation opportunities. Biophys. Rev. 2021, 13, 35–69. [Google Scholar] [CrossRef]
  60. Li, J.; Koh, J.-J.; Liu, S.; Lakshminarayanan, R.; Verma, C.S.; Beuerman, R.W. Membrane active antimicrobial peptides: Translating mechanistic insights to design. Front. Neurosci. 2017, 11, 73. [Google Scholar] [CrossRef] [Green Version]
  61. Mota-Meira, M.; LaPointe, G.; Lacroix, C.; Lavoie, M.C. MICs of mutacin B-Ny266, nisin A, vancomycin, and oxacillin against bacterial pathogens. Antimicrob. Agents Chemother. 2000, 44, 24–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Hoffmann, A.; Pag, U.; Wiedemann, I.; Sahl, H.G. Combination of antibiotic mechanisms in lantibiotics. Farmaco 2002, 57, 685–691. [Google Scholar] [CrossRef]
  63. Mygind, P.H.; Fischer, R.L.; Schnorr, K.M.; Hansen, M.T.; Sönksen, C.P.; Ludvigsen, S.; Raventós, D.; Buskov, S.; Christensen, B.; De Maria, L.; et al. Plectasin is a peptide antibiotic with therapeutic potential from a saprophytic fungus. Nature 2005, 437, 975–980. [Google Scholar] [CrossRef] [PubMed]
  64. Castiglione, F.; Cavaletti, L.; Losi, D.; Lazzarini, A.; Carrano, L.; Feroggio, M.; Ciciliato, I.; Corti, E.; Candiani, G.; Marinelli, F.; et al. A novel lantibiotic acting on bacterial cell wall synthesis produced by the uncommon actinomycete Planomonospora sp. Biochemistry 2007, 46, 5884–5895. [Google Scholar] [CrossRef] [PubMed]
  65. Bonelli, R.R.; Schneider, T.; Sahl, H.G.; Wiedemann, I. Insights into in vivo activities of lantibiotics from gallidermin and epidermin mode-of-action studies. Antimicrob. Agents Chemother. 2006, 50, 1449–1457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Castiglione, F.; Lazzarini, A.; Carrano, L.; Corti, E.; Ciciliato, I.; Gastaldo, L.; Candiani, P.; Losi, D.; Marinelli, F.; Selva, E.; et al. Determining the structure and mode of action of microbisporicin, a potent lantibiotic active against multiresistant pathogens. Chem. Biol. 2008, 15, 22–31. [Google Scholar] [CrossRef] [Green Version]
  67. Appleyard, A.N.; Choi, S.; Read, D.M.; Lightfoot, A.; Boakes, S.; Hoffmann, A.; Chopra, I.; Bierbaum, G.; Rudd, B.A.; Dawson, M.J.; et al. Dissecting structural and functional diversity of the lantibiotic mersacidin. Chem. Biol. 2009, 16, 490–498. [Google Scholar] [CrossRef] [Green Version]
  68. Piper, C.; Draper, L.A.; Cotter, P.D.; Ross, R.P.; Hill, C. A comparison of the activities of lacticin 3147 and nisin against drug-resistant Staphylococcus aureus and Enterococcus species. J. Antimicrob. Chemother. 2009, 64, 546–551. [Google Scholar] [CrossRef] [Green Version]
  69. Wescombe, P.A.; Heng, N.C.; Burton, J.P.; Chilcott, C.N.; Tagg, J.R. Streptococcal bacteriocins and the case for Streptococcus salivarius as model oral probiotics. Future Microbiol. 2009, 4, 819–835. [Google Scholar] [CrossRef] [Green Version]
  70. Oliynyk, I.; Varelogianni, G.; Roomans, G.M.; Johannesson, M. Effect of duramycin on chloride transport and intracellular calcium concentration in cystic fibrosis and non-cystic fibrosis epithelia. APMIS 2010, 118, 982–990. [Google Scholar] [CrossRef] [PubMed]
  71. Dawson, M.J.; Scott, R.W. New horizons for host defense peptides and lantibiotics. Curr. Opin. Pharmacol. 2012, 12, 545–550. [Google Scholar] [CrossRef] [Green Version]
  72. Kamarajan, P.; Hayami, T.; Matte, B.; Liu, Y.; Danciu, T.; Ramamoorthy, A.; Worden, F.; Kapila, S.; Kapila, Y. Nisin ZP, a bacteriocin and food preservative, inhibits head and neck cancer tumorigenesis and prolongs survival. PLoS ONE 2015, 10, e0131008. [Google Scholar] [CrossRef] [Green Version]
  73. Mohr, K.I.; Volz, C.; Jansen, R.; Wray, V.; Hoffmann, J.; Bernecker, S.; Wink, J.; Gerth, K.; Stadler, M.; Müller, R. Pinensins: The first antifungal lantibiotics. Angew. Chem. 2015, 54, 11254–11258. [Google Scholar] [CrossRef] [PubMed]
  74. Czyzewski, A.M.; Jenssen, H.; Fjell, C.D.; Waldbrook, M.; Chongsiriwatana, N.P.; Yuen, E.; Hancock, R.E.W.; Barron, A.E. In vivo, in vitro, and in silico characterization of peptoids as antimicrobial agents. PLoS ONE 2016, 11, e0135961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Mickels, N.; McManus, C.; Massaro, J.; Friden, P.; Braman, V.; D’Agostino, R.; Oppenheim, F.; Warbington, M.; Dibart, S.; Van Dyke, T. Clinical and microbial evaluation of a histatin-containing mouthrinse in humans with experimental gingivitis. J. Clin. Periodontol. 2001, 28, 404–410. [Google Scholar] [CrossRef]
  76. Kollef, M.; Pittet, D.; Sánchez García, M.; Chastre, J.; Fagon, J.Y.; Bonten, M.; Hyzy, R.; Fleming, T.R.; Fuchs, H.; Bellm, L.; et al. A randomized double-blind trial of iseganan in prevention of ventilator-associated pneumonia. Am. J. Respir. Crit. Med. 2006, 173, 91–97. [Google Scholar] [CrossRef] [PubMed]
  77. Elad, S.; Epstein, J.B.; Raber-Durlacher, J.; Donnelly, P.; Strahilevitz, J. The antimicrobial effect of Iseganan HCl oral solution in patients receiving stomatotoxic chemotherapy: Analysis from a multicenter, double-blind, placebo-controlled, randomized, phase III clinical trial. J. Oral Pathol. Med. 2012, 41, 229–234. [Google Scholar] [CrossRef] [PubMed]
  78. van der Velden, W.J.F.M.; van Iersel, T.M.P.; Blijlevens, N.M.A.; Donnelly, J.P. Safety and tolerability of the antimicrobial peptide human lactoferrin 1-11 (hLF1-11). BMC Med. 2009, 7, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Fjell, C.D.; Hiss, J.A.; Hancock, R.E.W.; Schneider, G. Designing antimicrobial peptides: Form follows function. Nat. Rev. Drug Discov. 2012, 11, 37–51. [Google Scholar] [CrossRef]
  80. Alam, M.Z.; Wu, X.; Mascio, C.; Chesnel, L.; Hurdle, J.G. Mode of action and bactericidal properties of surotomycin against growing and nongrowing Clostridium difficile. Antimicrob. Agents Chemother. 2015, 59, 5165–5170. [Google Scholar] [CrossRef] [Green Version]
  81. Nilsson, A.C.; Janson, H.; Wold, H.; Fugelli, A.; Andersson, K.; Håkangård, C.; Olsson, P.; Olsen, W.M. LTX-109 Is a novel agent for nasal decolonization of methicillin-resistant and -sensitive Staphylococcus aureus. Antimicrob. Agents Chemother. 2015, 59, 145. [Google Scholar] [CrossRef] [Green Version]
  82. Kudrimoti, M.; Curtis, A.; Azawi, S.; Worden, F.; Katz, S.; Adkins, D.; Bonomi, M.; Elder, J.; Sonis, S.T.; Straube, R.; et al. Dusquetide: A novel innate defense regulator demonstrating a significant and consistent reduction in the duration of oral mucositis in preclinical data and a randomized, placebo-controlled phase 2a clinical study. J. Biotechnol. 2016, 239, 115–125. [Google Scholar] [CrossRef] [Green Version]
  83. Ming, L.; Huang, J.A. The antibacterial effects of antimicrobial peptides OP-145 against clinically isolated multi-resistant strains. Jpn. J. Infect. Dis. 2017, 70, 601–603. [Google Scholar] [CrossRef] [Green Version]
  84. Guo, L.; Edlund, A. Targeted antimicrobial peptides: A novel technology to eradicate harmful Streptococcus mutans. J. Calif. Dent. Assoc. 2017, 45, 557–564. [Google Scholar]
  85. Dale, G.E.; Halabi, A.; Petersen-Sylla, M.; Wach, A.; Zwingelstein, C. Pharmacokinetics, tolerability, and safety of murepavadin, a novel antipseudomonal antibiotic, in subjects with mild, moderate, or severe renal function impairment. Antimicrob. Agents Chemother. 2018, 62, e00490-18. [Google Scholar] [CrossRef] [Green Version]
  86. Håkansson, J.; Ringstad, L.; Umerska, A.; Johansson, J.; Andersson, T.; Boge, L.; Rozenbaum, R.T.; Sharma, P.K.; Tollbäck, P.; Björn, C.; et al. Characterization of the in vitro, ex vivo, and in vivo efficacy of the antimicrobial peptide DPK-060 used for topical treatment. Front. Cell. Infect. Microbiol. 2019, 9, 174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wilson, A.P. Clinical pharmacokinetics of teicoplanin. Clin. Pharmacokinet. 2000, 39, 167–183. [Google Scholar] [CrossRef] [PubMed]
  88. Li, J.; Nation, R.L.; Turnidge, J.D.; Milne, R.W.; Coulthard, K.; Rayner, C.R.; Paterson, D.L. Colistin: The re-emerging antibiotic for multidrug-resistant Gram-negative bacterial infections. Lancet Infect. Dis. 2006, 6, 589–601. [Google Scholar] [CrossRef]
  89. Chen, A.Y.; Zervos, M.J.; Vazquez, J.A. Dalbavancin: A novel antimicrobial. Int. J. Clin. Pract. 2007, 61, 853–863. [Google Scholar] [CrossRef] [Green Version]
  90. Greenberg, M. Enfuvirtide: From basic science to FDA approval. In Entry Inhibitors in HIV Therapy; Birkhäuser: Basel, Switzerland, 2007; pp. 161–177. [Google Scholar]
  91. Saravolatz, L.D.; Stein, G.E.; Johnson, L.B. Telavancin: A novel lipoglycopeptide. Clin. Infect. Dis. 2009, 49, 1908–1914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Bouza, E.; Burillo, A. Oritavancin: A novel lipoglycopeptide active against Gram-positive pathogens including multiresistant strains. Int. J. Antimicrob. Agents 2010, 36, 401–407. [Google Scholar] [CrossRef] [Green Version]
  93. Lublin, A.; Isoda, F.; Patel, H.; Yen, K.; Nguyen, L.; Hajje, D.; Schwartz, M.; Mobbs, C. FDA-approved drugs that protect mammalian neurons from glucose toxicity slow aging dependent on cbp and protect against proteotoxicity. PLoS ONE 2011, 6, e27762. [Google Scholar] [CrossRef]
  94. Muir, A.J. Telaprevir for the treatment of chronic hepatitis C infection. Expert Rev. Anti Infect. Ther. 2011, 9, 1105–1114. [Google Scholar] [CrossRef] [PubMed]
  95. Moore, J.A.; Meakin, M.; Earl, M.H.; Kummer, T.M.; McAleer, J.P.; Long, T.E. Effects of caspofungin, tolcapone and other FDA-approved medications on MRSA susceptibility to vancomycin. J. Glob. Antimicrob. Resist. 2020, 22, 283–289. [Google Scholar] [CrossRef] [PubMed]
  96. Ingham, A.B.; Moore, R.J. Recombinant production of antimicrobial peptides in heterologous microbial systems. Biotechnol. Appl. Biochem. 2007, 47, 1–9. [Google Scholar] [CrossRef] [PubMed]
  97. Li, Y.; Chen, Z. RAPD: A database of recombinantly-produced antimicrobial peptides. FEMS Microbiol. Lett. 2008, 289, 126–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Rao, X.; Hu, J.; Li, S.; Jin, X.; Zhang, C.; Cong, Y.; Hu, X.; Tan, Y.; Huang, J.; Chen, Z.; et al. Design and expression of peptide antibiotic hPAB-beta as tandem multimers in Escherichia coli. Peptides 2005, 26, 721–729. [Google Scholar] [CrossRef] [PubMed]
  99. Xu, X.; Jin, F.; Yu, X.; Ji, S.; Wang, J.; Cheng, H.; Wang, C.; Zhang, W. Expression and purification of a recombinant antibacterial peptide, cecropin, from Escherichia coli. Protein Expr. Purif. 2007, 53, 293–301. [Google Scholar] [CrossRef] [PubMed]
  100. Gupta, S.K.; Shukla, P. Sophisticated cloning, fermentation, and purification technologies for an enhanced therapeutic protein production: A review. Front. Pharmacol. 2017, 8, 419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Parachin, N.S.; Mulder, K.C.; Viana, A.A.; Dias, S.C.; Franco, O.L. Expression systems for heterologous production of antimicrobial peptides. Peptides 2012, 38, 446–456. [Google Scholar] [CrossRef]
  102. Chatterjee, D.K.; Esposito, D. Enhanced soluble protein expression using two new fusion tags. Protein Expr. Purif. 2006, 46, 122–129. [Google Scholar] [CrossRef]
  103. Xu, Z.; Zhong, Z.; Huang, L.; Peng, L.; Wang, F.; Cen, P. High-level production of bioactive human beta-defensin-4 in Escherichia coli by soluble fusion expression. Appl. Microbiol. Biotechnol. 2006, 72, 471–479. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, Z.; Wang, D.; Cong, Y.; Wang, J.; Zhu, J.; Yang, J.; Hu, Z.; Hu, X.; Tan, Y.; Hu, F.; et al. Recombinant antimicrobial peptide hPAB-β expressed in Pichia pastoris, a potential agent active against methicillin-resistant Staphylococcus aureus. Appl. Microbiol. Biotechnol. 2011, 89, 281–291. [Google Scholar] [CrossRef]
  105. Cregg, J.M.; Tolstorukov, I.; Kusari, A.; Sunga, J.; Madden, K.; Chappell, T. Expression in the yeast Pichia pastoris. Methods Enzymol. 2009, 463, 169–189. [Google Scholar] [CrossRef]
  106. Holaskova, E.; Galuszka, P.; Frebort, I.; Oz, M.T. Antimicrobial peptide production and plant-based expression systems for medical and agricultural biotechnology. Biotechnol. Adv. 2015, 33, 1005–1023. [Google Scholar] [CrossRef] [PubMed]
  107. Parvathy, S.T. Engineering plants as platforms for production of vaccines. Am. J. Plant Sci. 2020, 11, 707–735. [Google Scholar] [CrossRef]
  108. Ma, J.K.C.; Barros, E.; Bock, R.; Christou, P.; Dale, P.J.; Dix, P.J.; Fischer, R.; Irwin, J.; Mahoney, R.; Pezzotti, M.; et al. Molecular farming for new drugs and vaccines. Current perspectives on the production of pharmaceuticals in transgenic plants. EMBO Rep. 2005, 6, 593–599. [Google Scholar] [CrossRef]
  109. Twyman, R.M.; Schillberg, S.; Fischer, R. Transgenic plants in the biopharmaceutical market. Expert Opin. Emerg. Drugs 2005, 10, 185–218. [Google Scholar] [CrossRef]
  110. Korban, S.S. Targeting and expression of antigenic proteins in transgenic plants for production of edible oral vaccines. In Vitro Cell. Dev. Biol. Plant 2002, 38, 231–236. [Google Scholar] [CrossRef]
  111. Govea-Alonso, D.O.; Rybicki, E.; Rosales-Mendoza, S. Plant-based vaccines as a global vaccination approach: Current perspectives. In Genetically Engineered Plants as a Source of Vaccines against Wide Spread Diseases; Springer: New York, NY, USA, 2014; pp. 265–280. [Google Scholar] [CrossRef]
  112. Sinha, R.; Shukla, P. Antimicrobial peptides: Recent insights on biotechnological interventions and future perspectives. Protein Pept. Lett. 2019, 26, 79–87. [Google Scholar] [CrossRef] [PubMed]
  113. Bednarska, N.G.; Wren, B.W.; Willcocks, S.J. The importance of the glycosylation of antimicrobial peptides: Natural and synthetic approaches. Drug Discov. Today 2017, 22, 919–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Shanmugaraj, B.; Malla, A.; Phoolcharoen, W. Emergence of novel coronavirus 2019-nCoV: Need for rapid vaccine and biologics development. Pathogens 2020, 9, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Barta, A.; Sommergruber, K.; Thompson, D.; Hartmuth, K.; Matzke, M.A.; Matzke, A.J.M. The expression of a nopaline synthase—Human growth hormone chimaeric gene in transformed tobacco and sunflower callus tissue. Plant Mol. Biol. 1986, 6, 347–357. [Google Scholar] [CrossRef] [PubMed]
  116. Hiatt, A.; Caffferkey, R.; Bowdish, K. Production of antibodies in transgenic plants. Nature 1989, 342, 76–78. [Google Scholar] [CrossRef] [PubMed]
  117. Ma, J.K.-C.; Drossard, J.; Lewis, D.; Altmann, F.; Boyle, J.; Christou, P.; Cole, T.; Dale, P.; van Dolleweerd, C.J.; Isitt, V.; et al. Regulatory approval and a first-in-human phase I clinical trial of a monoclonal antibody produced in transgenic tobacco plants. Plant Biotechnol. J. 2015, 13, 1106–1120. [Google Scholar] [CrossRef] [PubMed]
  118. Ward, B.J.; Gobeil, P.; Séguin, A.; Atkins, J.; Boulay, I.; Charbonneau, P.-Y.; Couture, M.; D’Aoust, M.-A.; Dhaliwall, J.; Finkle, C.; et al. Phase 1 randomized trial of a plant-derived virus-like particle vaccine for COVID-19. Nat. Med. 2021, 27, 1071–1078. [Google Scholar] [CrossRef] [PubMed]
  119. Rybicki, E.P. Plant-made vaccines for humans and animals. Plant Biotechnol. J. 2010, 8, 620–637. [Google Scholar] [CrossRef]
  120. Marsian, J.; Lomonossoff, G.P. Molecular pharming—VLPs made in plants. Curr. Opin. Biotechnol. 2016, 37, 201–206. [Google Scholar] [CrossRef] [Green Version]
  121. Schillberg, S.; Finnern, R. Plant molecular farming for the production of valuable proteins—Critical evaluation of achievements and future challenges. J. Plant Physiol. 2021, 258–259, 153359. [Google Scholar] [CrossRef]
  122. Stoger, E.; Fischer, R.; Moloney, M.; Ma, J.K.-C. Plant molecular pharming for the treatment of chronic and infectious diseases. Annu. Rev. Plant Biol. 2014, 65, 743–768. [Google Scholar] [CrossRef]
  123. Rosales-Mendoza, S.; Nieto-Gómez, R. Green therapeutic biocapsules: Using plant cells to orally deliver biopharmaceuticals. Trends Biotechnol. 2018, 36, 1054–1067. [Google Scholar] [CrossRef]
  124. Rybicki, E.P. Plant-made vaccines and reagents for the One Health initiative. Hum. Vaccin. Immunother. 2017, 13, 2912–2917. [Google Scholar] [CrossRef]
  125. Chan, H.-T.; Daniell, H. Plant-made oral vaccines against human infectious diseases—Are we there yet? Plant Biotechnol. J. 2015, 13, 1056–1070. [Google Scholar] [CrossRef] [Green Version]
  126. Schillberg, S.; Raven, N.; Spiegel, H.; Rasche, S.; Buntru, M. Critical analysis of the commercial potential of plants for the production of recombinant proteins. Front. Plant Sci. 2019, 10, 720. [Google Scholar] [CrossRef] [PubMed]
  127. Shanmugaraj, B.; Ramalingam, S. Plant expression platform for the production of recombinant pharmaceutical proteins. Austin J. Biotechnol. Bioeng. 2014, 1, 4. [Google Scholar]
  128. Shanmugaraj, B.; Bulaon, C.J.I.; Phoolcharoen, W. Plant molecular farming: A viable platform for recombinant biopharmaceutical production. Plants 2020, 9, 842. [Google Scholar] [CrossRef]
  129. Xu, J.; Towler, M.; Weathers, P.J. Platforms for plant-based protein production. Bioprocess. Plant In Vitro Syst. 2018, 509–548. [Google Scholar] [CrossRef]
  130. Burnett, M.J.B.; Burnett, A.C. Therapeutic recombinant protein production in plants: Challenges and opportunities. Plants People Planet 2020, 2, 121–132. [Google Scholar] [CrossRef]
  131. Xu, J.; Dolan, M.C.; Medrano, G.; Cramer, C.L.; Weathers, P.J. Green factory: Plants as bioproduction platforms for recombinant proteins. Biotechnol. Adv. 2012, 30, 1171–1184. [Google Scholar] [CrossRef]
  132. Moon, K.-B.; Park, J.-S.; Park, Y.-I.; Song, I.-J.; Lee, H.-J.; Cho, H.S.; Jeon, J.-H.; Kim, H.-S. Development of systems for the production of plant-derived biopharmaceuticals. Plants 2020, 9, 30. [Google Scholar] [CrossRef] [Green Version]
  133. Leite, M.L.; Sampaio, K.B.; Costa, F.F.; Franco, O.L.; Dias, S.C.; Cunha, N.B. Molecular farming of antimicrobial peptides: Available platforms and strategies for improving protein biosynthesis using modified virus vectors. An. Acad. Bras Cienc. 2019, 91, e20180124. [Google Scholar] [CrossRef]
  134. Boominathan, A.; Vanhoozer, S.; Basisty, N.; Powers, K.; Crampton, A.L.; Wang, X.; Friedricks, N.; Schilling, B.; Brand, M.D.; O’Connor, M.S. Stable nuclear expression of ATP8 and ATP6 genes rescues a mtDNA Complex V null mutant. Nucleic Acids Res. 2016, 44, 9342–9357. [Google Scholar] [CrossRef] [Green Version]
  135. Zhang, Y.; Zhang, A.; Li, X.; Lu, C. The role of chloroplast gene expression in plant responses to environmental stress. Int. J. Mol. Sci. 2020, 21, 6082. [Google Scholar] [CrossRef] [PubMed]
  136. Shahid, N.; Daniell, H. Plant-based oral vaccines against zoonotic and non-zoonotic diseases. Plant Biotechnol. J. 2016, 14, 2079–2099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Gelvin, S.B. Agrobacterium-mediated plant transformation: The biology behind the “gene-jockeying” tool. Microbiol. Mol. Biol. Rev. 2003, 67, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Lee, L.-Y.; Gelvin, S. T-DNA binary vectors and systems. Plant Physiol. 2008, 146, 325–332. [Google Scholar] [CrossRef] [Green Version]
  139. Gehl, C.; Li, G.; Serek, M. An efficient protocol for Agrobacterium-mediated transformation and regeneration of Campanula medium (Canterbury bells) based on leaf disc explants. Plant Cell Tissue Organ Cult. PCTOC 2020, 140, 635–645. [Google Scholar] [CrossRef]
  140. Fahad, S.; Khan, F.A.; Pandupuspitasari, N.S.; Ahmed, M.M.; Liao, Y.C.; Waheed, M.T.; Sameeullah, M.; Darkhshan; Hussain, S.; Saud, S.; et al. Recent developments in therapeutic protein expression technologies in plants. Biotechnol. Lett. 2015, 37, 265–279. [Google Scholar] [CrossRef]
  141. Chebolu, S.; Daniell, H. Chloroplast-derived vaccine antigens and biopharmaceuticals: Expression, folding, assembly and functionality. Curr. Top. Microbiol. Immunol. 2009, 332, 33–54. [Google Scholar] [CrossRef]
  142. Gorantala, J.; Grover, S.; Goel, D.; Rahi, A.; Jayadev Magani, S.K.; Chandra, S.; Bhatnagar, R. A plant based protective antigen [PA(dIV)] vaccine expressed in chloroplasts demonstrates protective immunity in mice against anthrax. Vaccine 2011, 29, 4521–4533. [Google Scholar] [CrossRef]
  143. Song, I.; Kang, Y.J.; Kim, D.H.; Kim, M.K.; Ko, K. Expression and in vitro function of anti-cancer mAbs in transgenic Arabidopsis thaliana. BMB Rep. 2020, 53, 229–233. [Google Scholar] [CrossRef]
  144. Park, S.H.; Ji, K.-Y.; Kim, H.M.; Ma, S.H.; Park, S.Y.; Do, J.H.; Oh, D.-B.; Kang, H.S.; Shim, J.S.; Joung, Y.H. Optimization of the human colorectal carcinoma antigen GA733-2 production in tobacco plants. Plant Biotechnol. Rep. 2021, 15, 55–67. [Google Scholar] [CrossRef]
  145. Meneguetti, B.T.; Machado, L.d.S.; Oshiro, K.G.N.; Nogueira, M.L.; Carvalho, C.M.E.; Franco, O.L. Antimicrobial peptides from fruits and their potential use as biotechnological tools—A review and outlook. Front. Microbiol. 2017, 7, 2136. [Google Scholar] [CrossRef] [Green Version]
  146. Verma, D.; Samson, N.P.; Koya, V.; Daniell, H. A protocol for expression of foreign genes in chloroplasts. Nat. Protoc. 2008, 3, 739–758. [Google Scholar] [CrossRef]
  147. Daniell, H.; Lin, C.S.; Yu, M.; Chang, W.J. Chloroplast genomes: Diversity, evolution, and applications in genetic engineering. Genome Biol. 2016, 17, 134. [Google Scholar] [CrossRef] [Green Version]
  148. Zoschke, R.; Bock, R. Chloroplast translation: Structural and functional organization, operational control, and regulation. Plant Cell 2018, 30, 745–770. [Google Scholar] [CrossRef] [Green Version]
  149. Jin, S.; Daniell, H. The engineered chloroplast genome just got smarter. Trends Plant Sci. 2015, 20, 622–640. [Google Scholar] [CrossRef] [Green Version]
  150. Daniell, H.; Jin, S.; Zhu, X.-G.; Gitzendanner, M.A.; Soltis, D.E.; Soltis, P.S. Green giant—A tiny chloroplast genome with mighty power to produce high-value proteins: History and phylogeny. Plant Biotechnol. J. 2021, 19, 430–447. [Google Scholar] [CrossRef]
  151. Zhang, B.; Shanmugaraj, B.; Daniell, H. Expression and functional evaluation of biopharmaceuticals made in plant chloroplasts. Curr. Opin. Chem. Biol. 2017, 38, 17–23. [Google Scholar] [CrossRef] [Green Version]
  152. Verma, D.; Daniell, H. Chloroplast vector systems for biotechnology applications. Plant Physiol. 2007, 145, 1129–1143. [Google Scholar] [CrossRef] [Green Version]
  153. Cody, W.B.; Scholthof, H.B. Plant virus vectors 3.0: Transitioning into synthetic genomics. Annu. Rev. Phytopathol. 2019, 57, 211–230. [Google Scholar] [CrossRef]
  154. Salazar-González, J.; Rosales-Mendoza, S.; Bañuelos-Hernandez, B. Viral vector-based expression strategies. In Genetically Engineered Plants Source of Vaccines Against Wide Spread Diseases; Springer: New York, NY, USA, 2014; pp. 43–60. [Google Scholar]
  155. Ariga, H.; Toki, S.; Ishibashi, K. Potato virus X vector-mediated DNA-free genome editing in plants. Plant Cell Physiol. 2020, 61, 1946–1953. [Google Scholar] [CrossRef]
  156. Mardanova, E.S.; Blokhina, E.A.; Tsybalova, L.M.; Peyret, H.; Lomonossoff, G.P.; Ravin, N.V. Efficient transient expression of recombinant proteins in plants by the novel pEff vector based on the genome of potato virus X. Front. Plant Sci. 2017, 8, 247. [Google Scholar] [CrossRef]
  157. Komarova, T.V.; Baschieri, S.; Donini, M.; Marusic, C.; Benvenuto, E.; Dorokhov, Y.L. Transient expression systems for plant-derived biopharmaceuticals. Expert Rev. Vaccines 2010, 9, 859–876. [Google Scholar] [CrossRef]
  158. Tiwari, S.; Verma, P.C.; Singh, P.K.; Tuli, R. Plants as bioreactors for the production of vaccine antigens. Biotechnol. Adv. 2009, 27, 449–467. [Google Scholar] [CrossRef]
  159. Shih, S.M.; Doran, P.M. Foreign protein production using plant cell and organ cultures: Advantages and limitations. Biotechnol. Adv. 2009, 27, 1036–1042. [Google Scholar] [CrossRef]
  160. Voinnet, O.; Rivas, S.; Mestre, P.; Baulcombe, D. An enhanced transient expression system in plants based on suppression of gene silencing by the p19 protein of tomato bushy stunt virus. Plant J. 2003, 33, 949–956. [Google Scholar] [CrossRef] [Green Version]
  161. Ghidey, M.; Islam, S.M.A.; Pruett, G.; Kearney, C.M. Making plants into cost-effective bioreactors for highly active antimicrobial peptides. New Biotechnol. 2020, 56, 63–70. [Google Scholar] [CrossRef]
  162. Yao, J.; Weng, Y.; Dickey, A.; Wang, K.Y. Plants as factories for human pharmaceuticals: Applications and challenges. Int. J. Mol. Sci. 2015, 16, 28549–28565. [Google Scholar] [CrossRef]
  163. Plasson, C.; Michel, R.; Lienard, D.; Saint-Jore-Dupas, C.; Sourrouille, C.; de March, G.G.; Gomord, V. Production of recombinant proteins in suspension-cultured plant cells. Methods Mol. Biol. 2009, 483, 145–161. [Google Scholar] [CrossRef]
  164. Raven, N.; Rasche, S.; Kuehn, C.; Anderlei, T.; Klöckner, W.; Schuster, F.; Henquet, M.; Bosch, D.; Büchs, J.; Fischer, R.; et al. Scaled-up manufacturing of recombinant antibodies produced by plant cells in a 200-L orbitally-shaken disposable bioreactor. Biotechnol. Bioeng. 2015, 112, 308–321. [Google Scholar] [CrossRef]
  165. Magy, B.; Tollet, J.; Laterre, R.; Boutry, M.; Navarre, C. Accumulation of secreted antibodies in plant cell cultures varies according to the isotype, host species and culture conditions. Plant Biotechnol. J. 2014, 12, 457–467. [Google Scholar] [CrossRef]
  166. Fox, J.L. First plant-made biologic approved. Nat. Biotechnol. 2012, 30, 472. [Google Scholar] [CrossRef]
  167. Schillberg, S.; Raven, N.; Fischer, R.; Twyman, R.M.; Schiermeyer, A. Molecular farming of pharmaceutical proteins using plant suspension cell and tissue cultures. Curr. Pharm. Des. 2013, 19, 5531–5542. [Google Scholar] [CrossRef]
  168. Santos, R.B.; Abranches, R.; Fischer, R.; Sack, M.; Holland, T. Putting the spotlight back on plant suspension cultures. Front. Plant Sci. 2016, 7. [Google Scholar] [CrossRef] [Green Version]
  169. Patiño-Rodríguez, O.; Ortega-Berlanga, B.; Llamas-González, Y.Y.; Flores-Valdez, M.A.; Herrera-Díaz, A.; Montes-de-Oca-Luna, R.; Korban, S.S.; Alpuche-Solís, Á.G. Transient expression and characterization of the antimicrobial peptide protegrin-1 in Nicotiana tabacum for control of bacterial and fungal mammalian pathogens. Plant Cell Tissue Organ Cult. PCTOC 2013, 115, 99–106. [Google Scholar] [CrossRef]
  170. Chahardoli, M.; Fazeli, A.; Ghabooli, M. Recombinant production of bovine Lactoferrin-derived antimicrobial peptide in tobacco hairy roots expression system. Plant Physiol. Biochem. 2018, 123, 414–421. [Google Scholar] [CrossRef]
  171. Chahardoli, M.; Fazeli, A.; Niazi, A.; Ghabooli, M. Recombinant expression of LFchimera antimicrobial peptide in a plant-based expression system and its antimicrobial activity against clinical and phytopathogenic bacteria. Biotechnol. Biotechnol. Equip. 2018, 32, 714–723. [Google Scholar] [CrossRef] [Green Version]
  172. Holásková, E.; Galuszka, P.; Micuchova, A.; Šebela, M.; Oz, T.; Frébort, I. Molecular farming in barley: Development of a novel production platform to produce human antimicrobial peptide LL-37. Biotechnol. J. 2018, 13, 1700628. [Google Scholar] [CrossRef]
  173. Wu, T.; Tang, D.; Chen, W.; Huang, H.; Wang, R.; Chen, Y. Expression of antimicrobial peptides thanatin(S) in transgenic Arabidopsis enhanced resistance to phytopathogenic fungi and bacteria. Gene 2013, 527, 235–242. [Google Scholar] [CrossRef]
  174. Jung, Y.J.; Lee, S.Y.; Moon, Y.S.; Kang, K.K. Enhanced resistance to bacterial and fungal pathogens by overexpression of a human cathelicidin antimicrobial peptide (hCAP18/LL-37) in Chinese cabbage. Plant Biotechnol. Rep. 2012, 6, 39–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Balaji, V.; Smart, C.D. Over-expression of snakin-2 and extensin-like protein genes restricts pathogen invasiveness and enhances tolerance to Clavibacter michiganensis subsp. michiganensis in transgenic tomato (Solanum lycopersicum). Transgenic Res. 2012, 21, 23–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Rong, W.; Qi, L.; Wang, J.; Du, L.; Xu, H.; Wang, A.; Zhang, Z. Expression of a potato antimicrobial peptide SN1 increases resistance to take-all pathogen Gaeumannomyces graminis var. tritici in transgenic wheat. Funct. Integr. Genom. 2013, 13, 403–409. [Google Scholar] [CrossRef]
  177. DeGray, G.; Rajasekaran, K.; Smith, F.; Sanford, J.; Daniell, H. Expression of an antimicrobial peptide via the chloroplast genome to control phytopathogenic bacteria and fungi. Plant Physiol. 2001, 127, 852–862. [Google Scholar] [CrossRef]
  178. Osusky, M.; Osuska, L.; Kay, W.; Misra, S. Genetic modification of potato against microbial diseases: In vitro and in planta activity of a dermaseptin B1 derivative, MsrA2. Theor. Appl. Genet. 2005, 111, 711–722. [Google Scholar] [CrossRef]
  179. Yevtushenko, D.P.; Misra, S. Comparison of pathogen-induced expression and efficacy of two amphibian antimicrobial peptides, MsrA2 and temporin A, for engineering wide-spectrum disease resistance in tobacco. Plant Biotechnol. J. 2007, 5, 720–734. [Google Scholar] [CrossRef] [Green Version]
  180. Chan, Y.-L.; Prasad, V.; Sanjaya; Chen, K.H.; Liu, P.C.; Chan, M.-T.; Cheng, C.-P. Transgenic tomato plants expressing an Arabidopsis thionin (Thi2.1) driven by fruit-inactive promoter battle against phytopathogenic attack. Planta 2005, 221, 386–393. [Google Scholar] [CrossRef]
  181. Prasad, B.D.; Jha, S.; Chattoo, B.B. Transgenic indica rice expressing Mirabilis jalapa antimicrobial protein (Mj-AMP2) shows enhanced resistance to the rice blast fungus Magnaporthe oryzae. Plant Sci. 2008, 175, 364–371. [Google Scholar] [CrossRef]
  182. Song, L.; Zhao, D.-g.; Wu, Y.-j.; Li, Y. Transient expression of chicken alpha interferon gene in lettuce. J. Zhejiang Univ. Sci. B 2008, 9, 351–355. [Google Scholar] [CrossRef]
  183. Sarowar, S.; Kim, Y.J.; Kim, K.D.; Hwang, B.K.; Ok, S.H.; Shin, J.S. Overexpression of Lipid Transfer Protein (LTP) genes enhances resistance to plant pathogens and LTP functions in long-distance systemic signaling in tobacco. Plant Cell Rep. 2009, 28, 419–427. [Google Scholar] [CrossRef]
  184. Jha, S.; Tank, H.G.; Prasad, B.D.; Chattoo, B.B. Expression of Dm-AMP1 in rice confers resistance to Magnaporthe oryzae and Rhizoctonia solani. Transgenic Res. 2009, 18, 59–69. [Google Scholar] [CrossRef] [PubMed]
  185. Lee, T.T.; Chang, C.C.; Juang, R.S.; Chen, R.B.; Yang, H.Y.; Chu, L.W.; Wang, S.R.; Tseng, T.H.; Wang, C.S.; Chen, L.J.; et al. Porcine lactoferrin expression in transgenic rice and its effects as a feed additive on early weaned piglets. J. Agric. Food Chem. 2010, 58, 5166–5173. [Google Scholar] [CrossRef] [PubMed]
  186. Jha, S.; Chattoo, B.B. Expression of a plant defensin in rice confers resistance to fungal phytopathogens. Transgenic Res. 2010, 19, 373–384. [Google Scholar] [CrossRef]
  187. Jan, P.-S.; Huang, H.-Y.; Chen, H.-M. Expression of a synthesized gene encoding cationic peptide cecropin B in transgenic tomato plants protects against bacterial diseases. Appl. Environ. Microbiol. 2010, 76, 769–775. [Google Scholar] [CrossRef] [Green Version]
  188. Lee, S.-B.; Li, B.; Jin, S.; Daniell, H. Expression and characterization of antimicrobial peptides retrocyclin-101 and protegrin-1 in chloroplasts to control viral and bacterial infections. Plant Biotechnol. J. 2011, 9, 100–115. [Google Scholar] [CrossRef] [Green Version]
  189. Ghag, S.B.; Shekhawat, U.K.; Ganapathi, T.R. Petunia floral defensins with unique prodomains as novel candidates for development of fusarium wilt resistance in transgenic banana plants. PLoS ONE 2012, 7, e39557. [Google Scholar] [CrossRef]
  190. Fukuta, S.; Kawamoto, K.-i.; Mizukami, Y.; Yoshimura, Y.; Ueda, J.-i.; Kanbe, M. Transgenic tobacco plants expressing antimicrobial peptide bovine lactoferricin show enhanced resistance to phytopathogens. Plant Biotechnol. 2012, 29, 383–389. [Google Scholar] [CrossRef] [Green Version]
  191. Verma, S.S.; Yajima, W.R.; Rahman, M.H.; Shah, S.; Liu, J.-J.; Ekramoddoullah, A.K.M.; Kav, N.N.V. A cysteine-rich antimicrobial peptide from Pinus monticola (PmAMP1) confers resistance to multiple fungal pathogens in canola (Brassica napus). Plant Mol. Biol. 2012, 79, 61–74. [Google Scholar] [CrossRef]
  192. Cabanos, C.; Ekyo, A.; Amari, Y.; Kato, N.; Kuroda, M.; Nagaoka, S.; Takaiwa, F.; Utsumi, S.; Maruyama, N. High-level production of lactostatin, a hypocholesterolemic peptide, in transgenic rice using soybean A1aB1b as carrier. Transgenic Res. 2013, 22, 621–629. [Google Scholar] [CrossRef]
  193. Zeitler, B.; Bernhard, A.; Meyer, H.; Sattler, M.; Koop, H.-U.; Lindermayr, C. Production of a de-novo designed antimicrobial peptide in Nicotiana benthamiana. Plant Mol. Biol. 2013, 81, 259–272. [Google Scholar] [CrossRef]
  194. Jung, Y.-J. Enhanced resistance to bacterial pathogen in transgenic tomato plants expressing cathelicidin antimicrobial peptide. Biotechnol. Bioprocess Eng. 2013, 18, 615–624. [Google Scholar] [CrossRef]
  195. Company, N.; Nadal, A.; La Paz, J.-L.; Martínez, S.; Rasche, S.; Schillberg, S.; Montesinos, E.; Pla, M. The production of recombinant cationic α-helical antimicrobial peptides in plant cells induces the formation of protein bodies derived from the endoplasmic reticulum. Plant Biotechnol. J. 2014, 12, 81–92. [Google Scholar] [CrossRef] [PubMed]
  196. Bundó, M.; Montesinos, L.; Izquierdo, E.; Campo, S.; Mieulet, D.; Guiderdoni, E.; Rossignol, M.; Badosa, E.; Montesinos, E.; San Segundo, B.; et al. Production of cecropin A antimicrobial peptide in rice seed endosperm. BMC Plant Biol. 2014, 14, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Schulz, S.; Stephan, A.; Hahn, S.; Bortesi, L.; Jarczowski, F.; Bettmann, U.; Paschke, A.-K.; Tusé, D.; Stahl, C.H.; Giritch, A.; et al. Broad and efficient control of major foodborne pathogenic strains of Escherichia coli by mixtures of plant-produced colicins. Proc. Natl. Acad. Sci. USA 2015, 112, E5454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Stephan, A.; Hahn-Löbmann, S.; Rosche, F.; Buchholz, M.; Giritch, A.; Gleba, Y. Simple purification of Nicotiana benthamiana-produced recombinant colicins: High-yield recovery of purified proteins with minimum alkaloid content supports the suitability of the host for manufacturing food additives. Int. J. Mol. Sci. 2017, 19, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Liu, Y.; Kamesh, A.C.; Xiao, Y.; Sun, V.; Hayes, M.; Daniell, H.; Koo, H. Topical delivery of low-cost protein drug candidates made in chloroplasts for biofilm disruption and uptake by oral epithelial cells. Biomaterials 2016, 105, 156–166. [Google Scholar] [CrossRef] [Green Version]
  200. Vetchinkina, E.M.; Komakhina, V.V.; Vysotskii, D.A.; Zaitsev, D.V.; Smirnov, A.N.; Babakov, A.V.; Komakhin, R.A. Expression of plant antimicrobial peptide pro-SmAMP2 gene increases resistance of transgenic potato plants to Alternaria and Fusarium pathogens. Genetika 2016, 52, 1055–1068. [Google Scholar] [CrossRef]
  201. Hao, G.; Zhang, S.; Stover, E. Transgenic expression of antimicrobial peptide D2A21 confers resistance to diseases incited by Pseudomonas syringae pv. tabaci and Xanthomonas citri, but not Candidatus Liberibacter asiaticus. PLoS ONE 2017, 12, e0186810. [Google Scholar] [CrossRef]
  202. Paškevičius, Š.; Starkevič, U.; Misiūnas, A.; Vitkauskienė, A.; Gleba, Y.; Ražanskienė, A. Plant-expressed pyocins for control of Pseudomonas aeruginosa. PLoS ONE 2017, 12, e0185782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Badrhadad, A.; Nazarian-Firouzabadi, F.; Ismaili, A. Fusion of a chitin-binding domain to an antibacterial peptide to enhance resistance to Fusarium solani in tobacco (Nicotiana tabacum). 3 Biotech 2018, 8, 391. [Google Scholar] [CrossRef]
  204. Shi, X.; Cordero, T.; Garrigues, S.; Marcos, J.F.; Daròs, J.-A.; Coca, M. Efficient production of antifungal proteins in plants using a new transient expression vector derived from tobacco mosaic virus. Plant Biotechnol. J. 2019, 17, 1069–1080. [Google Scholar] [CrossRef]
  205. Shams, M.V.; Nazarian-Firouzabadi, F.; Ismaili, A.; Shirzadian-Khorramabad, R. Production of a recombinant dermaseptin peptide in Nicotiana tabacum hairy roots with enhanced antimicrobial activity. Mol. Biotechnol. 2019, 61, 241–252. [Google Scholar] [CrossRef]
  206. Khademi, M.; Varasteh-Shams, M.; Nazarian-Firouzabadi, F.; Ismaili, A. New recombinant antimicrobial peptides confer resistance to fungal pathogens in tobacco plants. Front. Plant Sci. 2020, 11, 1236. [Google Scholar] [CrossRef] [PubMed]
  207. Łojewska, E.; Sakowicz, T.; Kowalczyk, A.; Konieczka, M.; Grzegorczyk, J.; Sitarek, P.; Skała, E.; Czarny, P.; Śliwiński, T.; Kowalczyk, T. Production of recombinant colicin M in Nicotiana tabacum plants and its antimicrobial activity. Plant Biotechnol. Rep. 2020, 14, 33–43. [Google Scholar] [CrossRef] [Green Version]
  208. MacDonald, J.; Doshi, K.; Dussault, M.; Hall, J.C.; Holbrook, L.; Jones, G.; Kaldis, A.; Klima, C.L.; Macdonald, P.; McAllister, T.; et al. Bringing plant-based veterinary vaccines to market: Managing regulatory and commercial hurdles. Biotechnol. Adv. 2015, 33, 1572–1581. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic representation of technologies available in plant-based platforms for the production of recombinant biopharmaceuticals. During initial stages of plant molecular farming, recombinant protein expression in plants were based on stable expression in which the gene of interest is cloned into plant expression vector and transformed into plant nucleus/chloroplast either by Agrobacterium or biolistic mediated method to produce transgenic or transplastomic plants. Transient expression combines the advantages of A. tumefaciens and plant viral elements for the rapid and high yield protein production. Genetically modified suspension-cultured plant cells integrate the advantages of whole-plant systems with those of bacterial fermentation and mammalian cell cultures. Suspension cultures can be grown in bioreactors under controlled environment for recombinant protein production.
Figure 1. Schematic representation of technologies available in plant-based platforms for the production of recombinant biopharmaceuticals. During initial stages of plant molecular farming, recombinant protein expression in plants were based on stable expression in which the gene of interest is cloned into plant expression vector and transformed into plant nucleus/chloroplast either by Agrobacterium or biolistic mediated method to produce transgenic or transplastomic plants. Transient expression combines the advantages of A. tumefaciens and plant viral elements for the rapid and high yield protein production. Genetically modified suspension-cultured plant cells integrate the advantages of whole-plant systems with those of bacterial fermentation and mammalian cell cultures. Suspension cultures can be grown in bioreactors under controlled environment for recombinant protein production.
Molecules 26 04032 g001
Table 2. Advantages and disadvantages of different host systems available for the production of heterologous proteins for pharmaceutical applications (adapted from Shanmugaraj et al. [114]).
Table 2. Advantages and disadvantages of different host systems available for the production of heterologous proteins for pharmaceutical applications (adapted from Shanmugaraj et al. [114]).
Expression SystemAdvantagesDisadvantages
Bacteria
  • Easy to manipulate
  • Low cost
  • High expression
  • Ease of scale up
  • Short turnaround time
  • Established regulatory procedures and approval
  • Improper folding
  • Lack of post-translational modifications, which may affect the protein function.
  • Endotoxin accumulation
Mammalian Cells
  • Proper folding and authentic post-translational modifications
  • Existing regulatory approval
  • High production cost
  • Expensive media and culture condition requirements
Yeast
  • Rapid growth and scalable
  • Easy to manipulate
  • Simple and inexpensive media requirements and culture conditions
  • Post-translational modifications of recombinant proteins
  • Difficulty in cell disruption due to the thick and hard cell walls
  • Hyperglycosylation of proteins
Insect cells
  • High expression levels
  • Ability to produce complex proteins including secreted, membrane and intracellular proteins
  • Proper folding and post-translational modifications
  • High cost and time consuming
  • Expensive media and culture condition requirements
Plants
  • Rapid and affordable
  • Optimized growth conditions
  • Free from pathogen and bacterial toxin contaminants
  • Economical
  • Post-translational modification somewhat similar like mammalian system
  • Regulatory compliance
  • Limited glycosylation capacity
Table 3. Some of the candidate AMPs expressed in plant hosts.
Table 3. Some of the candidate AMPs expressed in plant hosts.
Anti-Microbial PeptidePlant SpeciesStable or TransientNucleus or ChloroplastExpression LevelApplicationReference
MSI-99 (Magainin)Tobacco (Nicotiana tabacum)StableChloroplastUndefinedEnhanced resistance to phytopathogenic bacteria (Pseudomonas syringae) and fungi (Aspergillus flavus; Fusarium moniliforme; Verticillium dahlia)[177]
MsrA2 (Dermaseptin)Potato (Solanum tuberosum)StableNucleus1–5 μg/g FWBroad-range and enhanced resistance to virulent phytopathogenic fungi (Alternaria, Cercospora, Fusarium, Phytophthora, Pythium, Rhizoctonia; Verticillium sp.) and bacteria (Erwinia carotovora)[178]
Tobacco (Nicotiana tabacum)StableNucleus6–7 μg/g FWResistance to phytopathogenic fungi (Fusarium solani; F. oxysporum; Alternaria alternata; Botrytis cinerea; Sclerotinia sclerotiorum), oomycete (Pythium aphanidermatum) and bacterium (Pectobacterium carotovorum)[179]
Thi2.1 (Thionin)Tomato (Lycopersicon esculentum)StableNucleusUndefinedCrop protection
(F. oxysporum f. sp. lycopersici; R. solanacearum strain Pss4)
[180]
Mj-AMP2 (Knottin)Rice (Oryza sativa)StableNucleus0.32–0.38% total proteinEnhanced resistance to fungal pathogen (Magnaporthe oryzae)[181]
ChIFN-alpha (interferon-α)Lettuce (Lactuca sativa)TransientNucleus 0.393 μg/kg FWAntiviral activity against vesicular stomatitis virus (VSV)[182]
Lipid Transfer Proteins (LTPs)Tobacco (Nicotiana tabacum)StableNucleusUndefinedEnhanced resistance to pathogen (Phytophthora nicotianae; Pseudomonas syringae pv. tabaci)[183]
Dm-AMP1 (Defensin)Rice (Oryza sativa)StableNucleus0.43–0.57% total soluble proteinEnhanced resistance to pathogen (Magnaporthe oryzae; Rhizoctonia solani)[184]
rLF (Lactoferrin)Rice (Oryza sativa)StableNucleus0.1% rice bran weightFunctional feed additive on early weaned piglets [185]
Rs-AFP2 (Defensin)Rice (Oryza sativa)StableNucleus0.45–0.53% total soluble proteinEnhanced resistance to fungal pathogen (Magnaporthe oryzae; Rhizoctonia solani)[186]
CecB (Cecropin)Tomato (Solanum lycopersicum)StableNucleus0.001 µg/mg FWPlant protection against bacterial pathogens (Ralstonia solanacearum; Xanthomonas campestris)[187]
Retrocyclin-101 (Defensin)Tobacco (Nicotiana tabacum)StableChloroplast32–38% total soluble proteinControl viral (tobacco mosaic virus) and bacterial (Erwinia carotovora) infections[188]
Protegrin-1 (Cathelicidin)Tobacco (Nicotiana tabacum)StableChloroplast17–26% total soluble proteinControl bacterial infections (Erwinia carotovora) [188]
Tobacco (Nicotiana tabacum)TransientNucleusUndefinedControl mammalian bacteria (Klebsiella pneumoniae; Staphylococcus aureus; Escherichia coli; Mycobacterium bovis) and fungal (Candida albicans) pathogens[169]
Petunia Floral defensinsBanana (Musa spp.)StableNucleusUndefinedEffective resistance against pathogenic fungal Fusarium oxysporum f. sp. cubense (foc) infection[189]
Snakin-2 (Snakin)Tomato (Solanum lycopersicum)StableNucleusUndefinedEnhanced resistance to Clavibacter michiganensis subsp. michiganensis[175]
Lactoferricin B (Lactoferrin)Tobacco (Nicotiana tabacum)StableNucleusUndefinedEnhanced tolerance to pathogenic bacterial (Pseudomonas syringae pv. tabaci) and fungal (Botrytis cinerea) diseases[190]
PmAMP1 (cysteine-rich protein)Canola (Brassica napus)StableNucleusUndefinedEffective resistance against fungal pathogens (Alternaria brassicae; Leptosphaeria maculans; Sclerotinia sclerotiorum)[191]
hCAP18/LL-37 (Fusion of two cathelicidin antimicrobial proteins)Chinese cabbage (Brassica rapa cv. Osome)StableNucleusUndefinedEnhanced resistance to bacteria (P. carotovorum subsp. carotovorum) and fungal (Fusarium oxysporum f. sp. Lycopersici; Colletotrichum higginsianum; Rhizoctonia solani[174]
Lactostatin (anionic peptide)Rice (Oryza sativa)StableNucleus2 mg/g dry seedsAnti-hypercholestero lemic drug for potential clinical use[192]
SP1-1 (de-novo designed)Tobacco (Nicotiana benthamiana)TransientNucleus0.025 mg/g FWAntimicrobial activity (P. syringae pv. Syringae; P. syringae pv. Tomato; P. corrugate; Pectobacterium carotovorum ssp. carotovorum)[193]
SN-1 (Snakin)Wheat (Triticum aestivum)StableNucleusUndefinedAntifungal activity in vitro and enhanced resistance to fungus (Gaeumannomyces graminis var. tritici) and [176]
Thanatin (S) (synthetic thanatin)Arabidopsis (Arabidopsis thaliana)StableNucleusUndefinedAcquired resistance to bacterial pathogen (Pseudomonas syringae pv. tomato.) and fungal pathogens (Botrytis cinerea; powdery mildew)
Antibacterial and antifungal activity in vitro
[173]
LL-37 (Cathelicidin)Tomato (Solanum lycopersicum)StableNucleus16.8–58.2 µg/mL total soluble proteinEnhanced antibacterial activity (Pectobacterium carotovorum ssp. Carotovorum (Pcc); Xanthomonas campestris pv. Vesicatoria (Xcv)[194]
Barley (Hordeum vulgare L.)StableNucleus0.55 mg/kg seedsAntibacterial activity against E. coli TOP10 in vitro[172]
BP100.gtag (synthetic peptide)Rice (Oryza sativa)StableNucleus0.5% total soluble proteinPlant protection against bacterial pathogens (Erwinia amylovora; Pseudomonas syringae; Xanthomonas axonopodis)[195]
CecA (Cecropin)Rice (Oryza sativa)StableNucleus1–4 μg/g seedsResistance to fungal pathogen (Fusarium verticillioides) and bacterial pathogen (Dickeya dadantii)[196]
Recombinant colicins (Colicin)Tobacco (Nicotiana benthamiana)TransientNucleus0.6–3 mg/g FWEffective and broad control of foodborne pathogenic Escherichia coli strains [197]
Tobacco (Nicotiana benthamiana)TransientNucleus0.58–2.31 mg/g FWBroad activity, high potency, and purity as food antibacterial[198]
RetrocyclinTobaccoStableChloroplast116 μg of RC101/g of lyophilized leaf Effective against Streptococcus mutans and impaired biofilm formation following a single topical application of tooth-mimetic surface.[199]
ProtegrinTobaccoStableChloroplastUndefinedEffective against Streptococcus mutans and impaired biofilm formation following a single topical application of tooth-mimetic surface.[199]
pro-SmAMP2 (Hevein-like peptide)Potato (Solanum tuberosum)StableNucleusUndefinedCrop protection from Alternaria sp. and Fusarium sp. pathogens in resistant potato cultivar[200]
D2A21 (synthetic peptide)Citrus fruit (Carrizo citrange)StableNucleusUndefinedReduced development of canker disease caused by bacterium (Xanthomonas citri)[201]
PaeM4 (Pyocin)Tobacco (Nicotiana benthamiana)TransientNucleus800 μg/g FWBroad spectrum of antimicrobial activity against clinical isolates of Pseudomonas aeruginosa[202]
CBD-alfAFP (Defensin)Tobacco (Nicotiana tabacum)StableNucleusUndefinedEnhanced resistance to plant pathogen (Fusarium solani)[203]
LFchimera (Lactoferrin-derived peptides)Tobacco (Nicotiana tabacum)StableNucleusUndefinedAntimicrobial activity against clinical (Escherichia coli; Staphylococcus aureus) and phytopathogenic bacteria (Ralstonia solanacearum; Erwinia amylovira)[171]
Tobacco (Nicotiana tabacum)Suspension CulturesHairy roots4.8 μg/g FWEffective antimicrobial activity against Escherichia coli[170]
Penicillium digitatum AfpB (antifungal protein)Tobacco (Nicotiana benthamiana)TransientNucleus225 ± 37 µg/g FWProtect tomato plants against Botrytis cinerea causing grey mold disease [204]
DrsB1 (Dermaseptin)Tobacco (Nicotiana tabacum)Suspension cultureNucleusUndefinedEffective antimicrobial effects of plant bacterial and fungal phytopathogens [205]
Tobacco (Nicotiana tabacum)StableNucleus5.5–6.0 µg/g FWEnhanced resistance to plant pathogens (Alternaria alternata; Alternaria solani;, Fusarium oxysporum; Fusarium solani fungi)[206]
Laterosporulin-1 (synthetic anionic AMP/ELP fusion)Tobacco (Nicotiana benthamiana)TransientNucleus375 µg/g FWHigh antibacterial activity against Staphylococcus epidermidis[161]
ADP2-3 (synthetic anionic AMP/ELP fusion)Tobacco (Nicotiana benthamiana)TransientNucleus563 µg/g FWHigh antibacterial activity against Staphylococcus epidermidis[161]
Colicin M (Colicin)Tobacco (Nicotiana tabacum)StableNucleus2 mg/g FWAntibacterial activity against control and clinical pathogens (Escherichia coli; Klebsiella pneumoniae)[207]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shanmugaraj, B.; Bulaon, C.J.I.; Malla, A.; Phoolcharoen, W. Biotechnological Insights on the Expression and Production of Antimicrobial Peptides in Plants. Molecules 2021, 26, 4032. https://doi.org/10.3390/molecules26134032

AMA Style

Shanmugaraj B, Bulaon CJI, Malla A, Phoolcharoen W. Biotechnological Insights on the Expression and Production of Antimicrobial Peptides in Plants. Molecules. 2021; 26(13):4032. https://doi.org/10.3390/molecules26134032

Chicago/Turabian Style

Shanmugaraj, Balamurugan, Christine Joy I. Bulaon, Ashwini Malla, and Waranyoo Phoolcharoen. 2021. "Biotechnological Insights on the Expression and Production of Antimicrobial Peptides in Plants" Molecules 26, no. 13: 4032. https://doi.org/10.3390/molecules26134032

Article Metrics

Back to TopTop