Next Article in Journal
Identification of Colon Immune Cell Marker Genes Using Machine Learning Methods
Previous Article in Journal
Histopathological and Behavioral Impairments in Zebrafish (Danio rerio) Chronically Exposed to a Cocktail of Fipronil and Pyriproxyfen
Previous Article in Special Issue
Phytochemicals as Invaluable Sources of Potent Antimicrobial Agents to Combat Antibiotic Resistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antimicrobial Peptides: The Production of Novel Peptide-Based Therapeutics in Plant Systems

by
Pragya Tiwari
1,*,
Yashdeep Srivastava
2,
Abhishek Sharma
3 and
Ramachandran Vinayagam
1
1
Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
2
RR Institute of Modern Technology, Dr. A.P.J. Abdul Kalam Technical University, Sitapur Road, Lucknow 226201, Uttar Pradesh, India
3
Department of Biotechnology and Bioengineering, Institute of Advanced Research, Koba Institutional Area, Gandhinagar 392426, Gujarat, India
*
Author to whom correspondence should be addressed.
Life 2023, 13(9), 1875; https://doi.org/10.3390/life13091875
Submission received: 31 July 2023 / Revised: 25 August 2023 / Accepted: 4 September 2023 / Published: 7 September 2023

Abstract

:
The increased prevalence of antibiotic resistance is alarming and has a significant impact on the economies of emerging and underdeveloped nations. The redundancy of antibiotic discovery platforms (ADPs) and injudicious use of conventional antibiotics has severely impacted millions, across the globe. Potent antimicrobials from biological sources have been extensively explored as a ray of hope to counter the growing menace of antibiotic resistance in the population. Antimicrobial peptides (AMPs) are gaining momentum as powerful antimicrobial therapies to combat drug-resistant bacterial strains. The tremendous therapeutic potential of natural and synthesized AMPs as novel and potent antimicrobials is highlighted by their unique mode of action, as exemplified by multiple research initiatives. Recent advances and developments in antimicrobial discovery and research have increased our understanding of the structure, characteristics, and function of AMPs; nevertheless, knowledge gaps still need to be addressed before these therapeutic options can be fully exploited. This thematic article provides a comprehensive insight into the potential of AMPs as potent arsenals to counter drug-resistant pathogens, a historical overview and recent advances, and their efficient production in plants, defining novel upcoming trends in drug discovery and research. The advances in synthetic biology and plant-based expression systems for AMP production have defined new paradigms in the efficient production of potent antimicrobials in plant systems, a prospective approach to countering drug-resistant pathogens.

1. Overview of Global Antibiotic Resistance and Drug Discovery

The spread of antimicrobial resistance (AMR) has attained a state of ‘global emergency’ in the 21st century [1]. The statistics from the World Health Organization (WHO) have warned against the indiscriminate use of conventional antibiotics by humans in healthcare, the food sector, animal husbandry, and agriculture resulting in a high mortality of approximately 10 million deaths worldwide [2,3]. The rising frequency of antibiotic resistance and the emergence of drug-resistant diseases have a significant impact on developing countries’ economies [4,5], particularly those with limited healthcare facilities. The research initiatives have defined the ‘One Health and Global Health’ concept to address the critical themes in infectious diseases, particularly those contributing to AMR spread [6,7,8]. The interdisciplinary concept of ‘One Heath and Global Health’ provides knowledge-based information on the emergence of drug-resistant microbes and AMR evolution, the interconnected network of humans, animals, and the environment, and socio-economic factors in the spread of AMR [9,10]. Furthermore, studies have suggested that the presence of drug-resistant determinant factors in linked microbiomes alters bacterial genomes and triggers the prevalence of drug-resistant microbes [9]. An alarming rise in antibiotic resistance has been predicted by the WHO statistics suggesting 10 million mortalities every year by 2050, with most predicted causalities in the African and Asian sub-continent [11]. Besides the health of the people, the emergence of resistant microbes is worrisome in healthcare, particularly in surgical operations, transplants, and therapies for cancer treatment [12,13]. Alexander Fleming’s 1928 discovery of penicillin was a ‘phenomenal and pathbreaking discovery’ in the medical revolution, substantially expanding antibiotic arsenals for infection therapies [14]. The ‘antibiotic golden era’ was marked by streptomycin discovery and reached new paradigms with the subsequent development of sulfonamides, oxazolidinones, β-lactams antibiotics, cephalosporin, and tetracyclines, including others and the subsequently derived versions [15,16,17]. However, the efficacy and the usage of conventional antibiotics gradually declined, which is attributed to the development of drug resistance in multiple bacterial species.
The present trends in the rise of AMR can be ascribed to several causes, including indiscriminate/overuse, a lack of suitable marketing standards [18], and poor sanitation practices [19]. In the present context, methicillin-resistant Staphylococcus aureus (MRSA), Klebsiella pneumoniae, and Mycobacterium tuberculosis (extensively drug-resistant (XDR)) represent frequently present drug-resistant bacterial strains [20,21]. Some bacterial strains exhibited complete antibiotic resistance; for instance, colistin overuse (indiscriminate usage) to treat MDR strains (K. pneumonia, A. baumannii, and Pseudomonas aeruginosa) resulted in MCR-1-gene-mediated antibiotic resistance [22,23]. Although oxazolidinones and lipopeptides were marketed for Gram-negative microorganisms [24], staphylococcal resistance to oxazolidinones emerged subsequently [25]. The Food and Drug Administration (FDA) discontinued tigecycline and telithromycin due to their adverse effects [26,27]. M. tuberculosis, another bacterial strain, acquired fluoroquinolone resistance and other medicines (kanamycin, capreomycin, etc.) [28]. Daptomycin (lipopeptide) had some effectiveness in therapies but was subsequently withdrawn by the WHO [29]. The WHO produced a list of key MDR pathogens in 2017 that included third-generation cephalosporin-resistant P. aeruginosa, A. baumannii, and Enterobacteriaceae with an urgent necessity to tackle the emerging MDR strains [30].
The global rise in AMR further aggravated by chronic and multifactorial diseases is a threat to human and animal health and needs immediate attention. The development and characterization of novel antimicrobials from natural sources have contributed to the development of potent antimicrobials and their clinical trials in recent decades [31,32], but only a handful have shown promising outcomes. The research initiatives in this direction have explored the potential of antimicrobial peptides (AMPs) as antimicrobial agents, with key information deposited in AMP databases [33]. AMPs are emerging candidates as prospective antifungal and antibacterial therapies [34], attributed to their potential bioactivities. AMPs are a broad family of chemicals that are created as the first line of defense by multicellular organisms [35]. AMPs have been characterized as possible therapeutic alternatives to conventional therapy due to their minimal potential for resistance [36]. More than 3000 AMPs have been identified and described from multiple biological sources [37], including mammals [38], insects [39], amphibians [40], plants [41], and microorganisms [42]. In addition, AMPs are classified based on their bioactivities, namely antiparasitic, anti-tumor, antibacterial, antiviral, and anti-HIV, among others [43]. However, a detailed understanding of the AMP design and applications on the one hand and mechanisms of action on the other is crucial for creating or finding new AMPs with high biological efficacy against deadly microbial pathogens. Plants produce AMPs for a variety of purposes, including high-scale production via molecular farming and as a defense mechanism against multiple diseases (Figure 1).
The recent era has witnessed the emergence of plant systems as production platforms for AMPs as the preferred choice, attributed to synthesis in higher amounts, well-defined peptides (proper folding, glycosylation, etc.), and cost-effective peptide production, plants are favored for the manufacturing of AMPs [44]. Compared to bacterial and yeast expression systems, plant-based molecular farming is safer since there is essentially no chance of product contamination with infections that affect humans, animals, or endotoxins because plants do not have these pollutants. Substantial progress has been achieved in the production of AMPs in plant systems via synthetic biology [45]. The co-integration of synthetic biology strategies with system biology, genome engineering, and RNA interference (RNAi) for plant chassis and process optimization has yielded significant results in antimicrobial production [6,7], a plausible approach to addressing AMR. Through this thematic review, we herein present the recent trends in AMR prevalence and the prospects of AMPs as key arsenals to counter drug-resistant pathogens in a socio-economic context. In this direction, plants as production platforms and their genetic manipulation via synthetic biology comprise a sustainable approach to the production of novel and potent antimicrobials (Figure 2).

2. Natural Compounds and Derivatives as Potential Antimicrobial Agents

Natural compounds derived from plants, animals, and microbes are being extensively explored and studied for their antimicrobial properties [46,47,48]. Antibiotic-producing bacteria have been discovered in a variety of environmental niches, including extreme conditions (hypersaline locales) [49], plants (endophytes) [50], soil [51], marine sponges [52], and others. These antimicrobial compounds show a key potential to suppress the growth of bacteria [53] and fungi [54] and have major biotechnological/therapeutic uses. These bacteria demonstrate antimicrobial functions, and several antibiotics with high potency have been isolated from various microbial strains. Furthermore, several microbial strains were shown to suppress toxin synthesis and biofilm development [52,55]. Lactococcus lactis synthesizes a lantibiotic (nisin) that suppresses biofilm development in numerous species by eliminating or suppressing biofilm formation while having no negative effects on human beings. Moreira et al. demonstrated that boromycin, a macrolide from Streptomyces antibioticus, acts on transmembrane ion gradient and acts against non-growing and developing cells resistant to antibiotics. Plants constitute a rich reservoir of bioactive substances, and essential oils [24], triterpenoids [56], plant extracts [57], and plant proteins [58] have shown good efficacy in combating microbial infections in several animal models. Among plants, secondary metabolites with antimicrobial action are classified as phenolics, coumarins, saponins, alkaloids, terpenoids, and tannins [48,59] (Figure 3).
The enormous structural variation in chemical scaffolds of plant metabolites influences antibacterial activity, displaying multiple functions. For example, the –OH functional group found in phenolics inhibits microorganisms [59] by breaking the bacterial cell membrane and inducing cellular leakage [60], e.g., carvacrol. Eugenol is more efficient against Listeria and Campylobacter jejuni (due to double bond) than isoeugenol [61]. The antimicrobial mechanism of oxygenated terpenes comprises disruption of membranes and leakage of K+ ions [62]. Plant essential oils are potent antimicrobials, and limonene (1-methyl-4-(1-methyl ethenyl)-cyclohexene) is more effective than p-cymene in countering microbial infections [63]. In the case of terpenes, even minor structural changes in the location of the –OH group might drastically influence antibacterial action; for example, terpinene-4-ol at low concentrations promoted K+ leakage from E. coli cells when compared to terpineol. The capacity of oxygenated terpenes to break membranes and generate K+ leakage is responsible for the antibacterial effect [62]. Similarly, antibacterial activity was discovered in various plant by-products produced during food processing, in addition to other roles [64,65], due to its high phenolic content. As a result, the commercialization of by-products as antibacterials and maximal extraction would be significant in this direction.
Among the antimicrobial substances derived from animals, key examples include chitosan, lactoferrin, lactoperoxidase, lipids, and defensins [48]. Chitosan has sparked the interest of food preservationists; however, its usage is limited owing to higher and neutral pH insolubility and needs attention [66]. As acid-soluble chitosan substitutes, the chitosan derivatives have shown significant antimicrobial activity against B. cereus, E. coli, S. aureus, Shigella dysenteriae, and others [67]. Milk’s bioactive components (for example, casein) have been identified to have a variety of properties, one of which is antibacterial [68]. Coffee peels and husks [69], Bergamot peel (a by-product of essential oil) [70], pomegranate juice by-products [71], coconut husk [72], and others have antibacterial properties. Honey is a good source of bioactive substances and includes sugars, glycoproteins, etc., which have antibacterial action against MDR isolates [73]. Numerous additional bioactive compounds with antibacterial action have been identified from various natural resources, and a few notable examples are discussed (Table 1).

3. Antimicrobial Peptides: Novel Peptide-Based Therapeutics

AMPs are designated as a family of antibacterials that demonstrate multiple benefits including broad-spectrum anti-biofilm activity, delayed resistance development, and host immune response regulation [101]. AMPs are a class of tiny bioactive proteins that serve as a first line of defense against infections. AMPs’ mechanisms of action include immune response modulation, cell membrane disruption, and inflammation control [102] (Figure 4). AMPs are discovered in multiple niches, like soil and marine organisms, via different procedures, contributing to increased AMPs. Although 3,000 AMPs have been identified, only some have potent efficacy in clinical studies. Furthermore, the physicochemical characteristics of AMPs determine their antibacterial efficacy, absorption, and toxicity [101]. Studies have documented the safe application of synthetic AMPs in cattle, aquaculture, and as food preservatives, including the use of both natural and synthetic analogs of AMPs as antimicrobials [103,104].

3.1. Identification of AMPs from Nature and Properties

AMPs are biochemicals that have been conserved during evolution and are found in diverse organisms [105]. The primary characteristics of AMPs include (i) amphipathic nature (30% hydrophobic residues), (ii) positively charged molecules (+2 to +9 net positive charge) owing to positive amino acids such as lysine and arginine, and (iii) post-translational modification [106]. Furthermore, AMPs are encoded as one or more copies in biological genomes and are formed from bigger precursor molecules via post-translational modifications [106]. In the 1990s, the first AMPs were discovered in Drosophila melanogaster infected with bacteria or fungi [107]. A similar origin for Drosophila-produced human defensins has been suggested, whereas other AMPs (in Drosophila) exhibit homology with insect AMPs [108]. The co-evolution of AMPs in insects with other species emphasizes antimicrobial specificity, demonstrating synergistic effect and variation in specificity towards certain microbes. AMP has a broad spectrum of action and kills yeasts, bacteria, viruses, cancer cells, and fungi [109]. Plants and insects use AMPs to combat deadly microorganisms, whereas microbes use AMPs to preserve their habitats.
There are several secondary configurations for AMPs, including helices, disulfide bridges in strands, and extended/loop topologies that increase antimicrobial activity [110]. The AMPs are divided into cationic and non-cationic peptides (secretolytin) with varying structural characteristics (e.g., thanatin, penaeidins). Other characteristics include size, hydrophobicity, charge, size, self-association, etc., leading to broad-spectrum antibacterial activities [111]. Niyonsaba et al. [112] speculated that the co-evolution of AMPs may have been influenced by another characteristic of AMPs, namely the control of immune response by host cell contact. The mode of administration, target tissue, duration, dosage, and formulation, among other factors, affect how effective AMPs work against microbes. For instance, the production of AMP by the gut microbiota maintains bacterial colonization and reduces inflammation [113]. At the infection site, AMP may trigger the activation of innate immune cells and cause the induction of chemokines by a variety of cells [114]. Depending on the AMP type, it has different immunomodulatory features, including actions like those of growth factors and cytokines used to maintain immunological homeostasis [109].
The antimicrobial activity of AMPs is responsible for AMPs’ selectivity towards the target cells and is greatly influenced by their structural characteristics. Less than 100 hydrophobic and positively charged amino acid (lysine and arginine) residues make up an AMP molecule. The existence of AMPs in many biological species showed conservation over the course of evolution, and knowledge of the conserved structural features served as the foundation for creating new peptides [115]. Penaeidins, for instance, are chimeric cationic peptides that have antimicrobial action against fungi and Gram-positive bacteria. They have a chitin-binding domain (conserved), a PRP domain at the N-terminus, and a cysteine-rich domain at the C-terminus [116]. The AMP LL-37 is amphipathic and helical in shape and comprises XBBXBX patterns for heparin binding [117]. The physicochemical and structural characteristics of AMPs are another critical factor that greatly influences their toxicity toward certain cells. For instance, the AMP tachystatin was isolated from the horseshoe crab Tachypleus tridentatus and was antimicrobial towards fungal and bacterial strains. Tachystatin’s cytotoxic properties are thought to be caused by the presence of an amphiphilic sheet at the C-terminal end [118]. Antifungal peptides have not, however, been reported to include a conserved domain. Studies have focused on increasing AMP activity by using mutagenesis, peptide structural modification, or in silico methods. The capacity of AMPs to interact with cell membranes makes them a tumor-inducing/mitogenic agent [119] and a vector for the transport of drugs [120] and signaling molecules [121].

3.2. Classification and Discussion on the Structure of AMPs

In the ‘post-antibiotic era’, AMPs are emerging as potential antibacterial candidates and exhibit significant structural and functional diversity. As key components of innate immunity and the initial line of defense against microbial infections, AMPs are evolutionarily conserved in genomes. mRNA translation by ribosomes and nonribosomal peptide synthesis are the two methods that lead to the formation of AMPs in nature [122]. All living entities make and have genetically encoded AMPs from ribosomal translation, whereas non-ribosomally synthesized peptides originate from bacteria [122]. Ribosomally produced AMPs are increasingly employed as therapeutic agents for their immunological responses [35,62,109]. When AMPs are created as inactive precursors, proteases and their own expression are responsible for controlling how they are expressed [123].
According to the classification system for AMPs [124,125], most AMPs are classed as having β-sheet or α-helical structures. Despite not having a well-defined structure, the helical peptides bind with membranes to produce an amphipathic structure [126]. The two peptides from this category that have been examined as the most common are human lactoferrin and LL-97. Many AMPs are made up of β-sheet peptides, which are present in many kinds of plants, amphibians, marine invertebrates, etc. [127,128]. In the aqueous state, the β-sheet peptides have a rigid and well-organized structure and do not alter conformation as helical peptides. The antimicrobial properties of the β-sheet peptides include antibacterial, antiviral, antifungal, and anti-inflammatory effects [129]. Protegrins, defensins, and tachyplesins are some of the class’s more traditional members. The most studied β-sheet peptides are defensins, which are generated by neutrophils, epithelial cells, and macrophages as inactive precursor molecules [123,126]. These AMPs exhibit potent antibacterial action and are found in plants, invertebrates, and vertebrates [3]. Loops and extended-coil structures are included in the third class of AMPs. The extended-coil structure lacks β-sheets and -helices and is made up of amino acids such as proline, tryptophan, and arginine [35]. These AMPs work against Gram-negative bacteria by disrupting membranes and have anticancer effects as part of their broad-spectrum function [130]. Other significant examples are histatin (human saliva), tritrpticin, and indolicidin, a 13-amino acid peptide with strong antibacterial action [3,130].

3.3. Antimicrobial Peptides and Their Mechanism to Tackle Antibiotic Resistance

AMPs offer an advantage over conventional antibiotics since they are less prone to microbial resistance. These peptides act by disrupting membrane-bound pathogen activities and triggering the host immune system [131]. However, several issues must be resolved before AMP may be widely used as an antibiotic arsenal. Some related issues include production costs, toxicity parameters, and optimization (stability of AMPs). The development of new AMPs as therapeutic agents depends on significant research into the AMPs’ mechanism of action. Membrane-acting and non-membrane-acting AMPs are different types of AMPs. While non-membrane peptides pass across the membrane without disrupting it, membrane-acting AMPs are essentially cationic peptides that cause membrane disruption [132]. Antibacterial peptides that produce trans-membrane holes in the target membrane include LL-37 [133], defensins [134], and melittin [135]. Dermaseptin [136], buforin [137], and pleurocidin [138] are a few examples of AMPs that translocate across membranes (without producing disruption), among others. The transfer of these AMPs across cell membranes and the interruption of normal processes is their distinct mechanism of action [139]. Cell permeability and disruption are due to the cationic peptide’s interaction with the negatively charged lipopolysaccharide-containing cell membrane. Additionally, while interacting with microbial membranes, these AMPs display structural dynamics [140]. The other method of action involves blocking the creation of nucleic acids, proteins, and cell walls [141]. Additionally, AMP insertion into cell membranes is impacted by the fluidity of the membranes. The charge on the outer membrane, fluidity, etc., is essential for AMP membrane transport and influences AMP activity [142]. Additionally, certain AMPs target intracellular components by passing through the lipid bilayer and inhibit cellular processes including protein and nucleic acid production [143]. The mechanism of action of AMPs on bacterial membranes has been thoroughly studied in several models, including the carpet model [144], the toroidal pore model [145], and the barrel–stave mechanism [146].

3.4. AMPs as Antimicrobial Therapeutics: Clinical Validation and Trials

The AMPs highlight several benefits of their synthesis and use as antimicrobials, including high efficacy, negligible toxicity, and limited tissue accumulation. The AMPs are emerging pharmacological candidates, subject to clinical trials and validation, creating hope to expand their therapeutic applications [147]. The wider use of AMPs as therapeutics will be made possible by recent developments in peptide technology, including peptide drug conjugates, multifaceted peptides, and cell-penetrating peptides [148]. Globally, the United States produces and markets the most peptide-based medications, followed by Europe (Vicuron Pharmaceuticals and Theravance), which is principally engaged in peptide therapy research and development. Currently, over 60 peptide medications are commercially available, with additional new peptides undergoing clinical research and testing [149]. In 1999, magainin was discarded due to poor trial design, but there are still issues with peptide therapeutics that need to be resolved. One such peptide is MSI-78 which was unsuccessful in phase III clinical trials despite being effective against infections of diabetic foot ulcers. Three antimicrobial peptides (associated with indolicidin) were included in clinical trials [150]. MBI-226 was just recently launched and is currently in phase III of a clinical trial to treat catheter-related bloodstream infections [146]. By successfully reducing bacterial colonization in catheter-related infections [151] and showing antifungal activity against Candida albicans in guinea pigs [152], the clinical trials of MBI-226 have shown its efficiency in animal models. In a different study, Micrologix conducted phase II and III clinical trials for indolicidin-like peptides to treat acute acne and suppress MRSA [153]. AMPs and their derivatives have seen global commercial success in the treatment of infections [154]. The worldwide antimicrobial endeavors have given rise to extensive knowledge of AMPs’ effectiveness, mechanisms, safety, and other factors [155]. Additionally, greater funding for AMP research is needed to create peptide-based antibiotic drug arsenals.

4. Antimicrobial Peptide Production in Plants—Prospects and Advantages

Antimicrobial peptides (AMPs) comprise the first line of defense and comprise the integral component of immunity in all organisms. Recent years have witnessed increasing research on AMPs, which is attributed to their low toxicity to mammalian cells, unique action, and broad-spectrum functions [156]. The plant-based expression, also designated as molecular farming, offers a cost-effective approach to large-scale production of these therapeutic agents. Although the production of AMPs in plants is considered a prospective and emerging field of research in the present time, crucial challenges in terms of stability, function, and yield of the product account for some major concerns [157]. While plant-based expression systems are prospective as AMP production systems, an extended time is required for the optimization/generation of stable expression systems.

4.1. Plant Systems and Production of AMPs

Plants or animals are shielded from pathogenic attack by AMPs, serving as innate defense mechanisms [158]. There are currently more than 1700 known natural AMPs. Numerous derivatives and analogs have been created using computational systems or synthetically manufactured systems using natural AMPs as models. Because of advancements in biotechnology, it is now possible to produce proteins, peptides, and medicinal substances in large quantities using plants as bioreactors [156]. Plants produce AMPs for a variety of purposes, comprising large-scale synthesis in plants and plant protection from diseases. Due to well-organized peptides (correct folding, disulfide bond, and glycosylation) and cost-effective purification, plants are favored for the manufacturing of AMPs [43]. Additionally, compared to bacterial and yeast expression systems, plant-based molecular farming is safer since there is essentially no chance of product contamination with endotoxins or infections that affect humans or animals because plants do not have these pollutants. There are two main methods for producing AMPs in plants: steady integration of AMP genes into the plant genome, or a temporary transformation technique [159]. An illustration of molecular farming of antimicrobial peptides in plants is shown in Figure 2.

4.2. Plant Tissue Culture as Expression Systems

Tissue cultures, cell suspension cultures, callus cultures, and hairy root cultures are all methods used to sustain in vitro plants. Before creating a viable transgenic plant, the in vitro plant tissue culture approach is a strong tool for evaluating various expression processes [160]. This is so because plant cells or tissues may be grown in a controlled environment using regulated applications of phytohormones and culture media in in vitro cultures, which are independent of climatic variables. Agrobacterium rhizogenes is utilized in hairy root cultures in plant biotechnology to produce medicinal proteins and phytochemicals [161,162]. The platforms for molecular farming of antimicrobial peptides in plants are discussed. Every living plant tissue can be utilized to create a callus culture; however, young, meristematic tissues are the best sources of starting material [163]. Either direct or explant transformation of the callus and subsequent culture is used to create callus cultures that contain transgenes [164,165]. Monocotyledonous agricultural plants are typically resistant to in vitro regeneration and lack a screening platform for expression. As an alternate technique for screening stable transformants, callus cultures are used. With barley callus cultures, stable transgenics that express barley protein were created [166]. Hairy root cultures (HRCs) are the result of a plant becoming infected with the bacterium Rhizobium rhizogenes (initially A. rhizogenes). It may be generated from a variety of crops and allows the creation of extremely varied compounds [167]. HRCs have a steady biosynthetic capability and accumulate recombinant proteins in amounts comparable to those of entire plants. The stable integration of the gene of interest into the host plant genome is caused by root-inducing plasmids from A. rhizogenes [167]. A. rhizogenes was used to produce the chimeric antimicrobial peptides lactoferrampin and lactoferricin from bovine lactoferrin in tobacco hairy root cultures [44]. An appealing manufacturing strategy in terms of product consistency and the subsequent purification procedure is the expression of therapeutic proteins in in vitro plant systems under controlled circumstances [168]. ‘Next-generation human therapeutic antibodies’ are being created utilizing plant hairy root cultures [169,170,171]. Hairy root cultures of N. benthamiana are used to produce the tumor-targeting antibody-mAb H10 as part of an effort to discover next-generation therapeutic human antibodies [172].

4.3. Genetically Manipulated Plant Systems

Plant cells or tissues are used in plant molecular farming to express and process recombinant proteins or peptides with medicinal potential. This procedure is recognized as one of the most successful AMP manufacturing strategies [173]. Previously, potatoes and tobacco were commonly utilized as model plants for producing recombinant proteins, antigens, and medicines [174]. The two strategies that affect recombinant protein production in a plant are altering plant systems during synthesis (subcellular targeting, translation, and post-translational modifications) and controlling promoter-mediated transcription [175,176,177]. Constitutive production of the antimicrobial peptide cecropin P1 in transgenic potatoes resulted in plant resistance to the causal fungal pathogens of white rot and potato blight [178]. Most of the research employs constitutive expression of AMPs via ubiquitin and CaMV35S promoters [179,180,181]. Citrus Huanglongbing (HLB) and Candidatus Liberibacter asiaticus’ (Las) infections caused by Xanthomonas citri are the most damaging to citrus trees globally. Antimicrobial peptide D2A21 expression was performed to develop canker and HLB disease-resistant cultivars. Transgenic tobacco plants expressing D2A21 outperformed control plants in terms of disease resistance [182]. The LL-37 antibacterial peptide was also produced using transgenic barley. In a study, an alfalfa-plant-derived antifungal peptide was used to genetically modify potato plants [183]. Similarly, tachyplesin I, which offers defense against bacterial and fungal infections, was developed from the horseshoe crab [184]. In a different investigation, transgenic Ornithogalum plants expressed tachyplesin I [184]. The wild tobacco Nicotiana was modified for constitutive expression of the antimicrobial peptide (Mc-AMP1) from the common ice plant to examine the environmental and ecological implications of plant–microbe interactions. The outcome was that the transgenic plants displayed in-plant action against beneficial microorganisms for plants [185]. By using both direct and indirect transformation techniques, a Dermaseptin B1 recombinant antimicrobial peptide (C2-B1) was expressed in transgenic tobacco hairy root (HR) cultures. The total recombinant protein content of transgenic clones produced using direct and indirect transformation techniques and in liquid medium differs noticeably. According to the study, recombinant peptide production from the hairy root derived from the indirect transformation approach was noticeably greater [186]. Verticillium dahlia, an economically significant plant pathogen, is very resistant to the antimicrobial peptide BTD-S. Plants of the wild-type Columbia-0 ecotype of Arabidopsis thaliana expressed this peptide. In both in vivo and in vitro tests, BTD-S transgenic lines enhanced resistance toward V. dahliae [187]. The antimicrobial peptide PaDef (from Mexican avocado fruit) was heterologously expressed in Pichia pastoris, and the peptide restricted the development of S. aureus and E. coli [188] (Table 2).

4.4. Approaches for Transient Expression of AMPS

Due to its high expression levels, relative quickness, and high yield, transient transformation is a technology that is ideal for expressing a gene in plants and producing recombinant proteins [195]. The desired gene is expressed transiently in a plant host using a genetically designed vector, often Agrobacterium or a plant virus, and recombinant protein is obtained through expression within plant cells. This method allows for expression to begin within 24 h and continue for a week [196]. For temporary expression, plant viruses’ natural propensities for infection and Agrobacterium-mediated transformation are used. Entire plant leaves, plant suspension cells, or hairy root cultures are some of the components employed in temporary transformation [197]. Numerous peptides and proteins have been produced through temporary expression based on viral infection, including human interleukin-2, bovine lysozyme, human-galactosidase A, and bovine aprotinin, among others [198]. Using a TMV-based vector, the AMP recombinant aprotinin was created in tobacco plants. The second strategy involved employing a plant virus with high levels of expression and fast replication together with Agrobacterium to transfer DNA [199,200]. Heterologous proteins such as interferons, cytokines, growth hormones, etc., were produced using this method [187]. Vacuum penetration of Agrobacterium cultures changed tobacco plants. Even the creation of medicinal proteins or peptides for industries uses transient systems. Recombinant aprotinin was synthesized utilizing a Nicotiana sp. through a TMV-based vector in research. This work served as the initial illustration of the transient transformation technique for molecular farming’s generation of AMPs. The production of the avian H5N1 influenza vaccine in tobacco leaf tissues and its expression was carried out [201]. The Agrobacterium-mediated expression was used to synthesize the broad-spectrum AMP protegrin-1 (PG-1) in Nicotiana tabacum. According to Patio-Rodrguez et al. [202], this peptide was physiologically active against several bacterial and fungal species. The fruits of Raphanus sativus were resistant to bacterial spot disease by the induced production of SP1-1, an AMP targeted to the apoplast [203].
The use of plant cell suspension cultures requires consistent and specified conditions, which provide superior quality monitoring with a suitable medium, particularly during continuous manufacturing operations [195]. The scalability of plant cells, which is lower than that of full plants, is the main limitation of plant cell cultures. Additionally, it is not always possible to apply techniques that have been refined for plant cell cultures to actual plants [204]. Therefore, more screening and translational studies are further required. To establish a connection between plant cells and the scalability of complete plants, a plant cell pack (PCP), usually referred to as “cookies”, is created. The foundation of this platform is plant cell cultures without a liquid medium. Because of this, plant cell packs offer a viable platform for metabolic engineering, synthetic biology, and typical recombinant protein expression techniques that need high throughput screening of a variety of constructs for effective product development [205]. Human antibody chain proteins were molecularly farmed using PCP technology [206]. Rademacher developed a technique for the development and growth of a plant cell pack in 2020. A limitation to the industrial use of plant-based expression systems is the limited quantity of transiently expressed recombinant proteins generated via entire plants or plant cells. PCPs offer a quick and high-throughput screening approach with increased recombinant protein expression. In cell suspension cultures, namely A. thaliana, Catharanthus roseus, N. tabacum, etc., PCPs are effective. Due to fewer host proteins and secondary metabolites than in leaf-based expression systems, the approach also facilitates product purification. Agrobacterium tumefaciens-infused plant cell packs (PCPs), which are three-dimensional, porous aggregates of plant cells, were developed as an expression method [207]. For plant species like Nicotiana spp. and D. carota, these PCPs are more suited and effective than the transient expression approach in a liquid plant cell culture [206]. This uses both fast-growing cell suspension cultures and protein expression assays to understand vector replication efficiency detected as red and green fluorescent proteins [207]. The high-throughput screening of recombinant protein expression has been aided by the development of PCPs. The newly automated technique offers an affordable platform with more sample detection [208]. The same team of researchers has developed a technique for automatically transforming plant cell packs [208].

5. Computational Resources and Antimicrobial Research

The advancement of computational biology has made it easier to create vaccines and medicines with greater effectiveness. The development of antibacterials has centered on a combinational chemistry approach combined with computational modeling of the 3D structure of the antibacterial using energy minimization and other methods [209]. This approach aims to identify an effectively docked compound to a virulent gene, restricting the growth of the pathogen. The capacity to store proteomics/genomics findings and retrieve biological information has solved the issues connected with combating drug-resistant diseases. This has been made possible by the decoding of biological genomes and genome analysis using automated techniques [210]. The advancements in computational biology have made it easier to create vaccines and medicines with greater effectiveness. The antibacterial development has centered on combinational chemistry combined with computational modeling of the 3D structure of the lead compound using energy minimization and other techniques [209]. This approach aims to identify an effectively docked compound to a virulent gene, restricting the growth of the pathogen. The capacity to store proteomics/genomics findings and retrieve biological information has solved the issues connected with combating drug-resistant diseases. This has been made possible by the decoding of biological genomes and genome analysis using automated techniques [210]. Several bioinformatics databases and tools have also been created to give data on AMPs, information on microorganisms, and in silico methods to examine interactions between human pathogens.
Most of the research into the creation of antibacterial drugs has, up to this point, concentrated on the synthesis of lead candidates via inhibiting the synthesis of nucleic acids, limiting the biosynthesis of peptidoglycan, and blocking crucial metabolic pathways [210,211]. Through the identification of plasmid genes that inhibit the pathway, sequencing of genomes, and metabolic pathway reconstitution, computational biology has made a substantial contribution to drug development efforts [210]. To fully integrate bioinformatics and biochemical analysis for the generation of prospective drug candidates, key studies are still required. The advances in bioinformatics have contributed to our understanding of the existence of surface antigens and pathogenic genes. To create effective antimicrobial candidates for drug discovery and development, a deeper knowledge of microbial dynamics is required.

6. Bottlenecks in Research on AMPs as Antimicrobial Therapeutics

The substantial research and progress on AMPs, has necessitated the exploration of natural products to develop novel and potent candidates as antimicrobial agents. More than 3000 AMPs have been found and described to date [212], yet the bulk of them are unsuitable for treating humans (in their natural state) and many of them failed in clinical trials. AMPs, including telavancin, dalbavancin, vancomycin, daptomycin, oritavancin, and gramicidin D, have received FDA approval and are being marketed for use in medicine [213]. Many peptide-based therapies in the market function by activation or blocking of receptors, and others include pathway inhibitors and peptides that act on membranes. For usage as drug molecules, peptide-based treatments must be stable; most peptides authorized by the FDA exhibit stability in vivo [214]. Designing antimicrobials from bacteria and their biochemical aspects, combining AMPs with drugs for better function, and developing in vitro systems to study the antimicrobial effects are all necessary for creating synthetic AMPs [213]. Antimicrobial agents’ broad-spectrum action is caused structurally by the union of a clearly defined cationic region with a hydrophobic surface area, yet the rigidity of AMPs’ structure impairs their abilities to carry out the functions [128]. The main obstacles to AMP usage are its effectiveness and toxicity. Since some AMPs have in vivo side effects, they are exclusively employed in topical applications [215]. Additionally, the presence of serum and excessive salt levels might impact the antibacterial effects [144]. The vulnerability of AMPs to host proteolytic enzymes, which break down AMPs and influence their stability and pharmacokinetics, is another significant issue [216]. As a result, several AMPs are only recommended for use in tropical climates and are not recommended for oral administration. Current research has shown a more varied and complicated mechanism of AMPs, emphasizing a larger and multifaceted approach to combating microbial infections. In addition to their biological characteristics, additional elements such as in vivo stability [214], together with the resulting side effects and production costs, must be taken into consideration. To create AMPs with enhanced function, it will be crucial to have a thorough understanding of structure–function studies of AMPs and their effectiveness range.

7. Commercial Potential and Prospects of AMPs as Therapeutic Agents

AMPs are being developed as next-generation antibiotics to treat a variety of microbial illnesses, notably MDR strains, despite the numerous projected limitations. The tremendous therapeutic potential of AMPs, both natural and synthesized, as novel antimicrobials is highlighted by their multiple mechanisms of action. Our understanding of the structure and characteristics of AMPs has recently progressed, which is attributed to scientific advances, but there is still a long way to go before these therapeutic options can be fully utilized. Additionally, modified AMPs target or show specific antimicrobial activity with enhanced characteristics [217]. These modified AMPs include hybrid peptides, peptide mimetics, immobilized peptides, and peptide conjugates. These innovative peptides have potential uses in both industry and medicine, including as arsenals against antibiotic-resistant bacterial strains and for food preservation. AMR is on the rise all over the world, and AMPs have the potential to combat drug-resistant organisms, emphasizing the development of new antimicrobial options for viral, fungal, and bacterial illnesses. Short synthetic AMPs are frequently used to combat MDR microbial strains, and significant progress has recently been achieved in the design and synthesis of short peptides with increased effectiveness and decreased toxicity. The discovery of AMPs offers promising opportunities to replenish the depleting antibiotic pipeline and combat the global rise in AMR.

Author Contributions

Conceptualization, P.T.; writing—original draft preparation, P.T. and Y.S.; writing—review and editing, P.T., A.S. and R.V. 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

Not applicable.

Acknowledgments

The authors acknowledge their respective institutions for encouragement and support. We are grateful to the academic editor and reviewers for their valuable comments made for the improvement of the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Lack of New Antibiotics Threatens Global Efforts to Contain Drug-Resistant Infections. 2020. Available online: https://www.who.int/news/item/17-01-2020-lack-of-newantibiotics-threatens-global-efforts-to-contain-drug-resistant-infections (accessed on 11 December 2020).
  2. de Kraker, M.E.A.; Stewardson, A.J.; Harbarth, S. Will 10 million people die a year due to antimicrobial resistance by 2050? PLoS Med. 2016, 13, e1002184. [Google Scholar] [CrossRef] [PubMed]
  3. Mwangi, J.; Hao, X.; Lai, R.; Zhang, Z.-Y. Antimicrobial peptides: New hope in the war against multidrug resistance. Zool. Res. 2019, 40, 488–505. [Google Scholar] [CrossRef]
  4. Aslam, B.; Wang, W.; Arshad, M.I.; Khurshid, M.; Muzammil, S.; Rasool, M.H.; Nisar, M.A.; Alvi, R.F.; Aslam, M.A.; Qamar, M.U.; et al. Antibiotic resistance: A rundown of a global crisis. Infect. Drug Resist. 2018, 11, 1645–1658. [Google Scholar] [CrossRef] [PubMed]
  5. Zaman, S.B.; Hussain, M.A.; Nye, R.; Mehta, V.; Mamun, K.T.; Hossain, N. A review on antibiotic resistance: Alarm bells are ringing. Cureus 2017, 9, e1403. [Google Scholar] [CrossRef]
  6. Tiwari, P.; Khare, T.; Shriram, V.; Bae, H.; Kumar, V. Exploring synthetic biology strategies for producing potent antimicrobial phytochemicals. Biotechnol. Adv. 2021, 48, 107729. [Google Scholar] [CrossRef]
  7. Tiwari, P.; Srivastava, Y.; Kumar, V. Antimicrobial peptides as effective agents against drug-resistant pathogens. In Antimicrobial Resistance; Kumar, V., Shriram, V., Paul, A., Thakur, M., Eds.; Springer: Singapore, 2022; pp. 289–322. [Google Scholar]
  8. Tiwari, P.; Bajpai, M.; Sharma, A. Antimicrobials from medicinal plants: Key examples, success stories and prospects in tackling antibiotic resistance. Lett. Drug Des. Discov. 2023, 20, 420–438. [Google Scholar] [CrossRef]
  9. Hernando-Amado, S.; Coque, T.M.; Baquero, F.; Martínez, J.L. Defining and combating antibiotic resistance from One Health and Global Health perspectives. Nat. Microbiol. 2019, 4, 1432–1442. [Google Scholar] [CrossRef] [PubMed]
  10. Sharma, A.; Mistry, V.; Kumar, V.; Tiwari, P. Production of effective phyto-antimicrobials via metabolic engineering strategies. Curr. Top. Med. Chem. 2022, 22, 1068–1092. [Google Scholar] [CrossRef]
  11. O’Neill, J. Tackling Drug-Resistant Infections Globally: Final Report and Recommendations. The Review on Antimicrobial Resistance. 2016. Available online: https://amr-review.org/sites/default/files/160518_Final%20paper_with%20cover.pdf (accessed on 21 August 2023).
  12. Gudiol, C.; Carratalà, J. Antibiotic resistance in cancer patients. Expert Rev. Anti-Infect. Ther. 2014, 12, 1003–1016. [Google Scholar] [CrossRef]
  13. Lupei, M.I.; Mann, H.J.; Beilman, G.J.; Oancea, C.; Chipman, J.G. Inadequate antibiotic therapy in solid organ transplant recipients is associated with a higher mortality rate. Surg. Infect. 2010, 11, 33–39. [Google Scholar] [CrossRef]
  14. Van Epps, H.L. René Dubos: Unearthing antibiotics. J. Exp. Med. 2006, 203, 259. [Google Scholar] [CrossRef] [PubMed]
  15. Roncevic, T.; Vukicevic, D.; Krce, L.; Benincasa, M.; Aviani, I.; Maravic, A.; Tossi, A. Selection and redesign for high selectivity of membrane-active antimicrobial peptides from a dedicated sequence/function database. Biochim. Biophys. Acta Biomembr. 2019, 1861, 827–834. [Google Scholar] [CrossRef] [PubMed]
  16. Tiwari, P.; Srivastava, Y.; Bae, H. Trends of pharmaceutical design of Endophytes as anti-infective. Curr. Top. Med. Chem. 2021, 21, 1572–1586. [Google Scholar] [CrossRef]
  17. Tiwari, P.; Kang, S.; Bae, H. Plant-endophyte associations: Rich yet under-explored sources of novel bioactive molecules and applications. Microbiol. Res. 2023, 266, 127241. [Google Scholar] [CrossRef] [PubMed]
  18. Ayukekbong, J.A.; Ntemgwa, M.; Atabe, A.N. The threat of antimicrobial resistance in developing countries causes and control strategies. Antimicrob. Resist. Infect. Control 2017, 6, 47. [Google Scholar] [CrossRef]
  19. Davies, J.; Davies, D. Origins and evolution of antibiotic resistance. Microbiol. Mol. Biol. Rev. 2010, 74, 417–433. [Google Scholar] [CrossRef] [PubMed]
  20. Levin, A.S.; Barone, A.A.; Penço, J.; Santos, M.V.; Marinho, I.S.; Arruda, E.A.G.; Manrique, E.I.; Costa, S.F. Intravenous colistin as therapy for nosocomial infections caused by multidrug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii. Clin. Infect. Dis. 1999, 28, 1008–1011. [Google Scholar] [CrossRef]
  21. Miller, L.G.; Perdreau-Remington, F.; Rieg, G.; Mehdi, S.; Perlroth, J.; Bayer, A.S.; Tang, A.W.; Phung, T.O.; Spellberg, B. Necrotizing fasciitis caused by community-associated methicillin-resistant Staphylococcus aureus in Los Angeles. N. Engl. J. Med. 2005, 352, 1445–1453. [Google Scholar] [CrossRef]
  22. Liu, Y.Y.; Wang, Y.; Walsh, T.R.; Yi, L.X.; Zhang, R.; Spencer, J.; Doi, Y.; Tian, G.; Dong, B.; Huang, X.H.; et al. Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: A microbiological and molecular biological study. Lancet Infect. Dis. 2016, 16, 161–168. [Google Scholar] [CrossRef]
  23. Macnair, C.R.; Stokes, J.M.; Carfrae, L.A.; Fiebig-Comyn, A.A.; Coombes, B.K.; Mulvey, M.R.; Brown, E.D. Overcoming mcr-1 mediated colistin resistance with colistin in combination with other antibiotics. Nat. Commun. 2018, 9, 458. [Google Scholar] [CrossRef]
  24. Yu, Z.; Tang, J.; Khare, T.; Kumar, V. The alarming antimicrobial resistance in ESKAPEE pathogens: Can essential oils come to the rescue? Fitoterapia 2020, 140, 104433. [Google Scholar] [CrossRef]
  25. Eliopoulos, G.M.; Mek, V.G.; Gold, H.S. Antimicrobial resistance to Linezolid. Clin. Infect. Dis. 2004, 39, 1010–1015. [Google Scholar]
  26. Gleason, P.P.; Shaughnessy, A.F. Telithromycin (Ketek) for treatment of community-acquired pneumonia. Am. Fam. Physician 2007, 76, 1857. [Google Scholar]
  27. Dixit, D.; Madduri, R.P.; Sharma, R. The role of tigecycline in the treatment of infections in light of the new black box warning. Expert Rev. Anti-Infect. Ther. 2014, 12, 397–400. [Google Scholar] [CrossRef]
  28. Prestinaci, F.; Pezzotti, P.; Pantosti, A. Antimicrobial resistance: A global multifaceted phenomenon. Pathog. Glob. Health 2015, 109, 309–318. [Google Scholar] [CrossRef]
  29. World Health Organization. Executive summary: The selection and use of essential medicines in 2019. In Proceedings of the Report of the 22nd WHO Expert Committee on the Selection and Use of Essential Medicines, WHO, Headquarters, Geneva, Switzerland, 1–5 April 2019.
  30. WHO Global Priority List of Antibiotic-Resistant Bacteria to Guide Research, Discovery, and Development of New Antibiotics. Available online: http://www.who.int/medicines/publications/global-priority-listantibiotic-resistant-bacteria/en/ (accessed on 20 August 2023).
  31. Draenert, R.; Seybold, U.; Grützner, E.; Bogner, J.R. Novel antibiotics: Are we still in the pre-post-antibiotic era? Infection 2015, 43, 145–151. [Google Scholar] [CrossRef] [PubMed]
  32. Tiwari, P.; Bae, H. Endophytic fungi: Key insights, emerging prospects, and challenges in natural product drug discovery. Microorganisms 2022, 10, 360. [Google Scholar] [CrossRef] [PubMed]
  33. APD3 Antimicrobial Peptide Database, University of Nebraska Medical Center. Available online: https://aps.unmc.edu/ (accessed on 25 August 2023).
  34. Erdem Büyükkiraz, M.; Kesmen, Z. Antimicrobial peptides (AMPs): A promising class of antimicrobial compounds. J. Appl. Microbiol. 2022, 132, 1573–1596. [Google Scholar] [CrossRef] [PubMed]
  35. Seyfi, R.; Kahaki, F.A.; Ebrahimi, T.; Montazersaheb, S.; Eyvazi, S.; Babaeipour, V.; Tarhriz, V. Antimicrobial Peptides (AMPs): Roles, Functions and Mechanism of Action. Int. J. Pept. Res. Ther. 2020, 26, 1451–1463. [Google Scholar] [CrossRef]
  36. Mahlapuu, M.; Håkansson, J.; Ringstad, L.; Björn, C. Antimicrobial peptides: An emerging category of therapeutic agents. Front. Cell. Infect. Microbiol. 2016, 6, 194. [Google Scholar] [CrossRef] [PubMed]
  37. Ramazi, S.; Mohammadi, N.; Allahverdi, A.; Khalili, E.; Abdolmaleki, P. A review on antimicrobial peptides databases and the computational tools. Database 2022, 2022, baac011. [Google Scholar] [CrossRef]
  38. 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]
  39. 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]
  40. Conlon, J.M.; Mechkarska, M. Host-defense peptides with therapeutic potential from skin secretions of frogs from the family Pipidae. Pharmaceuticals 2014, 7, 58–77. [Google Scholar] [CrossRef]
  41. Tang, S.-S.; Prodhan, Z.H.; Biswas, S.K.; Le, C.-F.; Sekaran, S.D. Antimicrobial peptides from different plant sources: Isolation, characterization, and purification. Phytochemistry 2018, 154, 94–105. [Google Scholar] [CrossRef]
  42. Cao, J.; de la Fuente-Nunez, C.; Ou, R.W.; Torres, M.D.T.; Pande, S.G.; Sinskey, A.J.; Lu, T.K. Yeast-based synthetic biology platform for antimicrobial peptide production. ACS Synthet. Biol. 2018, 7, 896–902. [Google Scholar] [CrossRef]
  43. Huan, Y.; Kong, Q.; Mou, H.; Yi, H. Antimicrobial peptides: Classification, design, application and research progress in multiple fields. Front. Microbiol. 2020, 11, 582779. [Google Scholar] [CrossRef]
  44. Vriens, K.; Cammue, B.; Thevissen, K.J.M. Antifungal plant defensins: Mechanisms of action and production. Molecules 2014, 19, 12280–12303. [Google Scholar] [CrossRef]
  45. 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]
  46. Deshmukh, S.K.; Verekar, S.A.; Bhave, S.V. Endophytic fungi: A reservoir of antibacterials. Front. Microbiol. 2015, 5, 715. [Google Scholar] [CrossRef]
  47. da Silva, L.C.N.; da Silva, M.V.; Correia, M.T.D.S. Editorial: New frontiers in the search for antimicrobial agents from natural products. Front. Microbiol. 2017, 8, 210. [Google Scholar] [CrossRef]
  48. Gyawali, R.; Ibrahim, S.A. Natural products as antimicrobial agents. Food Control 2014, 46, 412–429. [Google Scholar] [CrossRef]
  49. Jose, P.A.; Jebakumar, S.R. Unexplored hypersaline habitats are sources of novel actinomycetes. Front. Microbiol. 2014, 5, 242. [Google Scholar] [CrossRef]
  50. Dos Santos, I.P.; da Silva, L.C.; da Silva, M.V.; de Araújo, J.M.; Cavalcanti, M.d.S.; Lima, V.L. Antibacterial activity of endophytic fungi from leaves of Indigofera suffruticosa Miller (Fabaceae). Front. Microbiol. 2015, 6, 350. [Google Scholar] [CrossRef]
  51. Hassan, R.; Shaaban, M.I.; Abdel Bar, F.M.; El-Mahdy, A.M.; Shokralla, S. Quorum sensing inhibiting activity of Streptomyces coelicoflavus isolated from soil. Front. Microbiol. 2016, 7, 659. [Google Scholar] [CrossRef]
  52. Saurav, K.; Bar-Shalom, R.; Haber, M.; Burgsdorf, I.; Oliviero, G.; Costantino, V.; Morgenstern, D.; Steindler, L. In search of alternative antibiotic drugs: Quorum-quenching activity in sponges and their bacterial isolates. Front. Microbiol. 2016, 7, 416. [Google Scholar] [CrossRef]
  53. de Oliveira, A.G.; Spago, F.R.; Simionato, A.S.; Navarro, M.O.; da Silva, C.S.; Barazetti, A.R.; Cely, M.V.; Tischer, C.A.; San Martin, J.A.; de Jesus Andrade, C.G.; et al. Bioactive Organocopper compound from Pseudomonas aeruginosa inhibits the growth of Xanthomonas citri subsp. citri. Front. Microbiol. 2016, 7, 113. [Google Scholar] [CrossRef]
  54. Kumar, V.; Naik, B.; Gusain, O.; Bisht, G.S. An actinomycete isolate from solitary wasp mud nest having strong antibacterial activity and kills the Candida cells due to the shrinkage and the cytosolic loss. Front. Microbiol. 2014, 5, 446. [Google Scholar] [CrossRef]
  55. Papa, R.; Selan, L.; Parrilli, E.; Tilotta, M.; Sannino, F.; Feller, G.; Tutino, M.L.; Artini, M. Anti-biofilm activities from marine cold-adapted bacteria against Staphylococci and Pseudomonas aeruginosa. Front. Microbiol. 2015, 6, 1333. [Google Scholar] [CrossRef]
  56. Wu, D.; Ding, W.; Zhang, Y.; Liu, X.; Yang, L. Oleanolic acid induces the type III secretion system of Ralstonia solanacearum. Front. Microbiol. 2015, 6, 1466. [Google Scholar] [CrossRef]
  57. Silva, A.P.S.A.; Silva, L.C.N.; Fonseca, C.S.M.; Araújo, J.M.; Santos Correia, M.T.; Silva Cavalcanti, M. Antimicrobial activity and phytochemical analysis of organic extracts from Cleome spinosa Jaqc. Front. Microbiol. 2016, 7, 963. [Google Scholar] [CrossRef]
  58. Patriota, L.L.; Procópio, T.F.; de Souza, M.F.; de Oliveira, A.P.; Carvalho, L.V.; Pitta, M.G.; Rego, M.J.; Paiva, P.M.; Pontual, E.V.; Napoleão, T.H. A Trypsin inhibitor from Tecoma stans leaves inhibits growth and promotes ATP depletion and lipid peroxidation in Candida albicans and Candida krusei. Front. Microbiol. 2016, 7, 611. [Google Scholar] [CrossRef]
  59. Cowan, M.M. Plant products as antimicrobial agents. Clin. Microbiol. Rev. 1999, 12, 564–582. [Google Scholar] [CrossRef]
  60. Xue, J.; Davidson, P.M.; Zhong, Q. Thymol nano-emulsified by whey protein-maltodextrin conjugates the enhanced emulsifying capacity and anti-listerial properties in milk by propylene glycol. J. Agric. Food Chem. 2013, 61, 12720–12726. [Google Scholar] [CrossRef]
  61. Friedman, M.; Henika, P.R.; Mandrell, R.E. Bactericidal activities of plant essential oils and some of their isolated constituents against Campylobacter jejuni, Escherichia coli, Listeria monocytogenes, and Salmonella enterica. J. Food Prot. 2002, 65, 1545–1560. [Google Scholar] [CrossRef]
  62. Griffin, S.G.; Wyllie, S.G.; Markham, J.L. Antimicrobially active terpenes cause Kþ leakage in E. coli cells. J. Essent. Oil Res. 2005, 17, 686–690. [Google Scholar] [CrossRef]
  63. Dorman, H.; Deans, S. Antimicrobial agents from plants: Antibacterial activity of plant volatile oils. J. Appl. Microbiol. 2000, 88, 308–316. [Google Scholar] [CrossRef]
  64. Tiwari, B.K.; Valdramidis, V.P.; O’Donnell, C.P.; Muthukumarappan, K.; Bourke, P.; Cullen, P. Application of natural antimicrobials for food preservation. J. Agric. Food Chem. 2009, 57, 5987–6000. [Google Scholar] [CrossRef]
  65. Balasundram, N.; Sundram, K.; Samman, S. Phenolic compounds in plants and agri-industrial by-products: Antioxidant activity, occurrence, and potential uses. Food Chem. 2006, 99, 191–203. [Google Scholar] [CrossRef]
  66. Du, Y.; Zhao, Y.; Dai, S.; Yang, B. Preparation of water-soluble chitosan from shrimp shell and its antibacterial activity. Innov. Food Sci. Emerg. Technol. 2009, 10, 103–107. [Google Scholar] [CrossRef]
  67. Chung, Y.C.; Yeh, J.Y.; Tsai, C.F. Antibacterial characteristics and activity of water-soluble chitosan derivatives prepared by the Maillard reaction. Molecules 2011, 16, 8504–8514. [Google Scholar] [CrossRef]
  68. Phelan, M.; Aherne, A.; FitzGerald, R.J.; O’Brien, N.M. Casein-derived bioactive peptides: Biological effects, industrial uses, safety aspects, and regulatory status. Int. Dairy J. 2009, 19, 643–654. [Google Scholar] [CrossRef]
  69. Esquivel, P.; Jimenez, V.M. Functional properties of coffee and coffee byproducts. Food Res. Int. 2012, 46, 488–495. [Google Scholar] [CrossRef]
  70. Mandalari, G.; Bennett, R.; Bisignano, G.; Trombetta, D.; Saija, A.; Faulds, C.; Gasson, M.; Narbad, A. Antimicrobial activity of flavonoids extracted from bergamot (Citrus bergamia Risso) peel, a byproduct of the essential oil industry. J. Appl. Microbiol. 2007, 103, 2056–2064. [Google Scholar] [CrossRef] [PubMed]
  71. Reddy, M.K.; Gupta, S.K.; Jacob, M.R.; Khan, S.I.; Ferreira, D. Antioxidant, antimalarial and antimicrobial activities of tannin-rich fractions, ellagitannins and phenolic acids from Punica granatum L. Planta Medica 2007, 73, 461–467. [Google Scholar] [CrossRef]
  72. Wonghirundecha, S.; Sumpavapol, P. Antibacterial activity of selected plant by-products against food-borne pathogenic bacteria. IPCBEE 2012, 39, 116–120. [Google Scholar]
  73. Brudzynski, K.; Sjaarda, C.; Lannigan, R. MRJP1-containing glycoproteins isolated from honey, a novel antibacterial drug candidate with broad spectrum activity against multi-drug resistant clinical isolates. Front. Microbiol. 2015, 6, 711. [Google Scholar] [CrossRef]
  74. Sakanaka, S.; Shimura, N.; Alzawa, M.; Kim, M.; Yamamoto, T. Preventive effect of green tea polyphenols against dental caries in conventional rats. Biosci. Biotechnol. Biochem. 1992, 56, 592–594. [Google Scholar] [CrossRef]
  75. Vijaya, K.; Ananthan, S.; Nalini, R. Antibacterial effect of theaflavin, polyphenon 60 (Camellia sinensis) and Euphorbia hirta on Shigella spp.-a cell culture study. J. Ethnopharmacol. 1995, 49, 115–118. [Google Scholar] [CrossRef]
  76. Kocacaliskan, I.; Talan, I.; Terzi, Z. Antimicrobial activity of catechol and pyrogallol as allelochemicals. Z. Naturforsch. C J. Biosci. 2006, 61, 639–642. [Google Scholar] [CrossRef]
  77. Ferdes, M. Antimicrobial compounds from plants. In Fighting Antimicrobial Resistance; Budimir, A., Ed.; Antimicrobial Compounds from Plants; IAPC-OBP: Zagreb, Croatia, 2018; pp. 271–273. [Google Scholar]
  78. Ignatowicz, E.; Baerdubowska, A.; Ignatowicz, W. Resveratrol, a natural chemopreventive agent against degenerative diseases. Pol. J. Pharmacol. 2001, 53, 557–569. [Google Scholar] [PubMed]
  79. Sun, D.; Hurdle, J.G.; Lee, R.; Cushman, M.; Pezzuto, J.M. Evaluation of flavonoid and resveratrol chemical libraries reveals Abyssinone II as a promising antibacterial lead. Chem. Med. Chem. 2012, 7, 1541–1545. [Google Scholar] [CrossRef] [PubMed]
  80. Su, Y.; Ma, L.; Wen, Y.; Wang, H.; Zhang, S. Studies of the in vitro antibacterial activities of several polyphenols against clinical isolates of methicillin-resistant Staphylococcus aureus. Molecules 2014, 19, 12630–12639. [Google Scholar] [CrossRef]
  81. Chovanova, R.; Mezovska, J.; Vaverkova, S.; Mikulasova, M. The inhibition of the Tet(K) efflux pump of tetracycline-resistant Staphylococcus epidermidis by essential oils from three Salvia species. Lett. Appl. Microbiol. 2015, 61, 58–62. [Google Scholar] [CrossRef]
  82. Dahlem Junior, M.A.; Nguema Edzang, R.W.; Catto, A.L.; Raimundo, J.-M. Quinones as an Efficient Molecular Scaffold in the Antibacterial/Antifungal or Antitumoral Arsenal. Int. J. Mol. Sci. 2022, 23, 14108. [Google Scholar] [CrossRef] [PubMed]
  83. Limaverde, P.W.; Campina, F.F.; da Cunha, F.A.; Crispim, F.D.; Figueredo, F.G.; Lima, L.F.; Oliveira-Tintino, C.D.d.M.; de Matos, Y.M.L.S.; Morais-Braga, M.F.B.; Menezes, I.R.A.; et al. Inhibition of the TetK efflux pump by the essential oil of Chenopodium ambrosioides L. and a-terpinene against Staphylococcus aureus IS-58. Food Chem. Toxicol. 2017, 109, 957–961. [Google Scholar] [CrossRef] [PubMed]
  84. Kalia, N.P.; Mahajan, P.; Mehra, R.; Nargotra, A.; Sharma, J.P.; Koul, S.; Khan, I.A. Capsaicin, a novel inhibitor of the NorA efflux pump, reduces the intracellular invasion of Staphylococcus aureus. J. Antimicrob. Chemother. 2012, 67, 2401–2408. [Google Scholar] [CrossRef]
  85. Malmir, M.; Serrano, R.; Duarte Silva, O.M. Anthraquinones as potential antimicrobial agents—A review. In Antimicrobial Research: Novel Bioknowledge and Educational Programs; Mendez-Vilas, A., Ed.; Formatex Research Center S.L.: Badajoz, Spain, 2017; ISBN 13: 9788494751202. [Google Scholar]
  86. Dwivedi, G.R.; Tyagi, R.; Tripathi, S.; Pati, S.; Srivastava, S.K.; Darokar, M.P.; Sharma, A. Antibiotics potentiating the potential of catharanthine against superbug Pseudomonas aeruginosa. J. Biomol. Struct. Dyn. 2018, 36, 4270–4284. [Google Scholar] [CrossRef]
  87. Park, C.; Woo, E.R.; Lee, D.G. Anti-Candida property of a lignan glycoside derived from Styrax japonica S. et Z. via membrane-active mechanisms. Mol. Cells 2010, 29, 581–584. [Google Scholar] [CrossRef]
  88. Chan, B.C.; Ip, M.; Lau, C.B.; Lui, S.L.; Jolivalt, C.; Ganem-Elbaz, C.; Litaudon, M.; Reiner, N.E.; Gong, H.; See, R.H.; et al. Synergistic effects of baicalein with ciprofloxacin against NorA over-expressed methicillin-resistant Staphylococcus aureus (MRSA) and inhibition of MRSA pyruvate kinase. J. Ethnopharmacol. 2011, 137, 767–773. [Google Scholar] [CrossRef]
  89. Dwivedi, G.; Maurya, A.; Yadav, D.; Khan, F.; Darokar, M.; Srivastava, S. Drug resistance reversal potential of ursolic acid derivatives against nalidixic acid- and multidrug-resistant Escherichia coli. Chem. Biol. Drug Des. 2014, 86, 272–283. [Google Scholar] [CrossRef] [PubMed]
  90. Oliveira, I.; Sousa, A.; Ferreira, I.C.; Bento, A.; Estevinho, L.; Pereira, J.A. Total phenols, antioxidant potential and antimicrobial activity of walnut (Juglans regia L.) green husks. Food Chem. Toxicol. 2008, 46, 2326–2331. [Google Scholar] [CrossRef]
  91. Sagdic, O.; Ozturk, I.; Yilmaz, M.T.; Yetim, H. Effect of grape pomace extracts obtained from different grape varieties on microbial quality of beef patty. J. Food Sci. 2011, 76, M515–M521. [Google Scholar] [CrossRef]
  92. Agourram, A.; Ghirardello, D.; Rantsiou, K.; Zeppa, G.; Belviso, S.; Romane, A.; Oufdou, K.; Giordano, M. Phenolic content, antioxidant potential, and antimicrobial activities of fruit and vegetable by-product extracts. Int. J. Food Propert. 2013, 16, 1092–1104. [Google Scholar] [CrossRef]
  93. Li, G.; Xu, Y.; Wang, X.; Zhang, B.; Shi, C.; Zhang, W.; Xia, X. Tannin-rich fraction from Pomegranate Rind damages membrane of Listeria monocytogenes. Foodborne Pathog. Dis. 2014, 11, 1–7. [Google Scholar] [CrossRef]
  94. Friedman, M.; Henika, P.R.; Levin, C.E. Bactericidal activities of health-promoting, food-derived powders against the foodborne pathogens Escherichia coli, Listeria monocytogenes, Salmonella enterica, and Staphylococcus aureus. J. Food Sci. 2013, 78, M270–M275. [Google Scholar] [CrossRef]
  95. Kazimierska, K.; Kalinowska-Lis, U. Milk proteins—Their biological activities and use in Cosmetics and Dermatology. Molecules 2021, 26, 3253. [Google Scholar] [CrossRef] [PubMed]
  96. Lucera, A.; Costa, C.; Conte, A.; Del Nobile, M.A. Food applications of natural antimicrobial compounds. Front. Microbiol. 2012, 3, 287. [Google Scholar] [CrossRef]
  97. Arques, J.L.; Fernandez, J.; Gaya, P.; Nunez, M.; Rodrıguez, E.; Medina, M. Antimicrobial activity of Reuterin in combination with nisin against food-borne pathogens. Int. J. Food Microbiol. 2004, 95, 225–229. [Google Scholar] [CrossRef]
  98. Eom, S.H.; Kim, Y.M.; Kim, S.K. Antimicrobial effect of phlorotannins from marine brown algae. Food Chem. Toxicol. 2012, 50, 3251–3255. [Google Scholar] [CrossRef] [PubMed]
  99. Bala, N.; Aitken, E.A.; Cusack, A.; Steadman, K.J. Antimicrobial potential of Australian macrofungi extracts against foodborne and other pathogens. Phytother. Res. 2012, 26, 465–469. [Google Scholar] [CrossRef]
  100. Oztürk, M.; Duru, M.E.; Kivrak, S.; Mercan-Dogan, N.; Türkoglu, A.; Ozler, M.A. In vitro antioxidant, anticholinesterase and antimicrobial activity studies on three Agaricus species with fatty acid compositions and iron contents: A comparative study on the three most edible mushrooms. Food Chem. Toxicol. 2011, 49, 1353–1360. [Google Scholar] [CrossRef] [PubMed]
  101. Magana, M.; Pushpanathan, M.; Santos, A.L.; Leanse, L.; Fernandez, M.; Ioannidis, A.; Giulianotti, M.A.; Apidianakis, Y.; Bradfute, S.; Ferguson, A.L.; et al. The value of antimicrobial peptides in the age of resistance. Lancet Infect. Dis. 2020, 20, e216–e230. [Google Scholar] [CrossRef]
  102. Mookherjee, N.; Hancock, R.E. Cationic host defense peptides: Innate immune regulatory peptides as a novel approach for treating infections. Cell. Mol. Life Sci. 2007, 64, 922–933. [Google Scholar] [CrossRef]
  103. Liu, Q.; Yao, S.; Chen, Y. Use of antimicrobial peptides as a feed additive for juvenile goats. Sci. Rep. 2017, 7, 12254. [Google Scholar] [CrossRef] [PubMed]
  104. Choi, S.C.; Ingale, S.L.; Kim, J.S.; Park, Y.K.; Kwon, I.K.; Chae, B.J. An antimicrobial peptide-A3: Effects on growth performance, nutrient retention, intestinal and fecal microflora, and intestinal morphology of broilers. Br. Poult. Sci. 2013, 54, 738–746. [Google Scholar] [CrossRef] [PubMed]
  105. Hancock, R.E. Cationic antimicrobial peptides: Towards clinical applications. Expert Opin. Investig. Drugs 2000, 9, 1723–1729. [Google Scholar] [CrossRef]
  106. Huerta-Cantillo, J.; Navarro-García, F. Properties and design of antimicrobial peptides as potential tools against pathogens and malignant cells. Investig. Discapac. 2016, 5, 96–115. [Google Scholar]
  107. Lemaitre, B.; Nicolas, E.; Michaut, L.; Reichhart, J.M.; Hoffmann, J.A. The dorsoventral regulatory gene cassette spätzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell 1996, 86, 973–983. [Google Scholar] [CrossRef]
  108. Mylonakis, E.; Podsiadlowski, L.; Muhammed, M.; Vilcinskas, A. Diversity, evolution and medical applications of insect antimicrobial peptides. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2016, 371, 371. [Google Scholar] [CrossRef]
  109. Zhang, L.J.; Gallo, R.L. Antimicrobial peptides. Curr. Biol. 2016, 26, R1–R21. [Google Scholar] [CrossRef] [PubMed]
  110. Hancock, R.E. Cationic peptides: Effectors in innate immunity and novel antimicrobials. Lancet Infect. Dis. 2001, 1, 156–164. [Google Scholar] [CrossRef]
  111. Pushpanathan, M.; Gunasekaran, P.; Rajendhran, J. Antimicrobial peptides: Versatile biological properties. Int. J. Pept. 2013, 675391, 1–15. [Google Scholar] [CrossRef] [PubMed]
  112. Niyonsaba, F.; Song, P.; Yue, H. Antimicrobial peptide derived from insulin-like growth factor-binding protein 5 activates mast cells via Mas-related G protein-coupled receptor X2. Allergy 2020, 75, 203–207. [Google Scholar] [CrossRef]
  113. Lhocine, N.; Ribeiro, P.S.; Buchon, N. PIMS modulates immune tolerance by negatively regulating Drosophila innate immune signaling. Cell Host Microbe 2008, 4, 147–158. [Google Scholar] [CrossRef]
  114. Hancock, R.E.; Haney, E.F.; Gill, E.E. The immunology of host defense peptides: Beyond antimicrobial activity. Nat. Rev. Immunol. 2016, 16, 321–334. [Google Scholar] [CrossRef]
  115. Yount, N.Y.; Yeaman, M.R. Multidimensional signatures in antimicrobial peptides. Proc. Natl. Acad. Sci. USA 2004, 101, 7363–7368. [Google Scholar] [CrossRef] [PubMed]
  116. Tassanakajon, A.; Amparyup, P.; Somboonwiwat, K.; Supungul, P. Cationic antimicrobial peptides in penaeid shrimp. Mar. Biotechnol. 2010, 12, 487–505. [Google Scholar] [CrossRef] [PubMed]
  117. Andersson, E.; Rydengard, V.; Sonesson, A.; Morgelin, M.; Bjorck, L.; Schmidtchen, A. Antimicrobial activities of heparin-binding peptides. Eur. J. Biochem. 2004, 271, 1219–1226. [Google Scholar] [CrossRef]
  118. Osaki, T.; Omotezako, M.; Nagayama, R. Horseshoe crab hemocyte-derived antimicrobial polypeptides, tachystatins, with sequence similarity to spider neurotoxins. J. Biol. Chem. 1999, 274, 26172–26178. [Google Scholar] [CrossRef]
  119. Utsugi, T.; Schroit, A.J.; Connor, J.; Bucana, C.D.; Fidler, I.J. Elevated expression of phosphatidylserine in the outer membrane leaflet of human tumor cells and recognition by activated human blood monocytes. Cancer Res. 1991, 51, 3062–3066. [Google Scholar] [PubMed]
  120. Henriques, S.T.; Melo, M.N.; Castanho, M.A.R.B. Cell-penetrating peptides, and antimicrobial peptides: How different are they? Biochem. J. 2006, 399, 1–7. [Google Scholar] [CrossRef] [PubMed]
  121. Blomqvist, M.; Bergquist, J.; Westman, A. Identification of defensins in human lymphocyte nuclei. Eur. J. Biochem. 1999, 263, 312–318. [Google Scholar] [CrossRef]
  122. Hancock, R.E.; Chapple, D.S. Peptide antibiotics. Antimicrob. Agents Chemother. 1999, 43, 1317–1323. [Google Scholar] [CrossRef] [PubMed]
  123. Lai, Y.; Gallo, R.L. AMPed up immunity: How antimicrobial peptides have multiple roles in immune defense. Trends Immunol. 2009, 30, 131–141. [Google Scholar] [CrossRef] [PubMed]
  124. Takahashi, D.; Shukla, S.K.; Prakash, O.; Zhang, G. Structural determinants of host defense peptides for antimicrobial activity and target cell selectivity. Biochimie 2010, 92, 1236–1241. [Google Scholar] [CrossRef]
  125. Lombardi, L.; Falanga, A.; Del Genio, V.; Galdiero, S. A new hope: Self-assembling peptides with antimicrobial activity. Pharmaceutics 2019, 11, 166. [Google Scholar] [CrossRef] [PubMed]
  126. Pasupuleti, M.; Schmidtchen, A.; Malmsten, M. Antimicrobial peptides: Key components of the innate immune system. Crit. Rev. Biotechnol. 2012, 32, 143–171. [Google Scholar] [CrossRef]
  127. Oppenheim, J.J.; Biragyn, A.; Kwak, L.W.; Yang, D. Roles of antimicrobial peptides such as defensins in innate and adaptive immunity. Ann. Rheum. Dis. 2003, 62, 17–21. [Google Scholar] [CrossRef] [PubMed]
  128. Nguyen, L.T.; Haney, E.F.; Vogel, H.J. The expanding scope of antimicrobial peptide structures and their modes of action. Trends Biotechnol. 2011, 29, 464–472. [Google Scholar] [CrossRef]
  129. Kumar, P.; Kizhakkedathu, J.N.; Straus, S.K. Antimicrobial peptides: Diversity, mechanism of action and strategies to improve the activity and biocompatibility in vivo. Biomolecules 2018, 8, 4. [Google Scholar] [CrossRef] [PubMed]
  130. Falla, T.J.; Karunaratne, D.N.; Hancock, R.E.W. Mode of action of the antimicrobial peptide indolicidin. J. Biol. Chem. 1996, 271, 19298–19303. [Google Scholar] [CrossRef] [PubMed]
  131. Lin, L.; Chi, J.; Yan, Y.; Luo, R.; Feng, X.; Zheng, Y.; Xian, D.; Li, X.; Quan, G.; Liu, D.; et al. Membrane-disruptive peptides/peptidomimetics-based therapeutics: Promising systems to combat bacteria and cancer in the drug-resistant era. Acta Pharm. Sin. B 2021, 11, 2609–2644. [Google Scholar] [CrossRef]
  132. Hancock, R.E.W.; Patrzykat, A. Clinical development of cationic antimicrobial peptides: From natural to novel antibiotics. Curr. Drug Targets Infect. Disord. 2002, 2, 79–83. [Google Scholar] [CrossRef] [PubMed]
  133. Xhindoli, D.; Pacor, S.; Benincasa, M.; Scocchi, M.; Gennaro, R.; Tossi, A. The human cathelicidin LL-37-A pore-forming antibacterial peptide and host-cell modulator. Biochim. Biophys. Acta 2016, 1858, 546–566. [Google Scholar] [CrossRef] [PubMed]
  134. Shafee, T.M.; Lay, F.T.; Phan, T.K.; Anderson, M.A.; Hulett, M.D. Convergent evolution of defensin sequence, structure, and function. Cell. Mol. Life Sci. 2017, 74, 663–682. [Google Scholar] [CrossRef] [PubMed]
  135. Sun, D.; Forsman, J.; Woodward, C.E. Molecular simulations of melittin-induced membrane pores. J. Phys. Chem. B 2017, 121, 10209–10214. [Google Scholar] [CrossRef] [PubMed]
  136. Belmadani, A.; Semlali, A.; Rouabhia, M. Dermaseptin-S1 decreases Candida albicans growth, biofilm formation, and the expression of hyphal wall protein 1 and aspartic protease genes. J. App. Microbiol. 2018, 125, 72–83. [Google Scholar] [CrossRef]
  137. Perez, R.H.; Ishibashi, N.; Inoue, T.; Himeno, K.; Masuda, Y.; Sawa, N. Functional analysis of genes involved in the biosynthesis of Enterocin NKR-5-3B, a novel circular bacteriocin. J. Bacteriol. 2016, 198, 291–300. [Google Scholar] [CrossRef]
  138. Zhang, M.; Wei, W.; Sun, Y.; Jiang, X.; Ying, X.; Tao, R.; Ni, L. Pleurocidin congeners demonstrate activity against Streptococcus and low toxicity on gingival fibroblasts. Arch. Oral Biol. 2016, 70, 79–87. [Google Scholar] [CrossRef]
  139. Lohner, K. Membrane-active antimicrobial peptides as template structures for novel antibiotic agents. Curr. Top. Med. Chem. 2017, 17, 508–519. [Google Scholar] [CrossRef] [PubMed]
  140. Mingeot-Leclercq, M.P.; Décout, J.L. Bacterial lipid membranes as promising targets to fight antimicrobial resistance, molecular foundations and illustration through the renewal of aminoglycoside antibiotics and the emergence of amphiphilic aminoglycosides. Med. Chem. Comm. 2016, 7, 586–611. [Google Scholar] [CrossRef]
  141. Cudic, M.; Otvos, L., Jr. Intracellular targets of antibacterial peptides. Curr. Drug Targets 2002, 3, 101–106. [Google Scholar] [CrossRef] [PubMed]
  142. Claro, B.; Bastos, M.; Garcia-Fandino, R. Design and applications of cyclic peptides. In Peptide Applications in Biomedicine, Biotechnology, and Bioengineering; Koutsopoulos, S., Ed.; Woodhead Publishing: Sawston, UK, 2018; pp. 87–129. [Google Scholar]
  143. Falanga, A.; Galdiero, S. Emerging therapeutic agents on the basis of naturally occurring antimicrobial peptides. In Amino Acids, Peptides Proteins; Ryadnov, M., Hudecz, F., Eds.; Royal Society of Chemistry: London, UK, 2017; Volume 42, pp. 190–227. [Google Scholar]
  144. Han, J.; Zhao, S.; Ma, Z.; Gao, L.; Li, H.; Muhammad, U. The antibacterial activity and modes of LI–F type antimicrobial peptides against Bacillus cereus in vitro. J. Appl. Microbiol. 2017, 123, 602–614. [Google Scholar] [CrossRef] [PubMed]
  145. Phoenix, D.A.; Dennison, S.R.; Harris, F. Bacterial resistance to host defense peptides. In Host Defense Peptides and Their Potential as Therapeutic Agents; Epand, R.M., Ed.; Springer: Cham, Switzerland, 2016; pp. 161–204. [Google Scholar]
  146. Shabir, U.; Ali, S.; Magray, A.R.; Ganai, B.A.; Firdous, P.; Hassan, T.; Nazir, R. Fish antimicrobial peptides (AMPs) as essential and promising molecular therapeutic agents: A review. Microb. Pathog. 2018, 114, 50–56. [Google Scholar] [CrossRef]
  147. 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]
  148. Raucher, D.; Ryu, J.S. Cell-penetrating peptides: Strategies for anticancer treatment. Trends Mol. Med. 2015, 21, 560–570. [Google Scholar] [CrossRef]
  149. Lau, J.L.; Dunn, M.K. Therapeutic peptides: Historical perspectives, current development trends, and future directions. Bioorg. Med. Chem. 2018, 26, 2700–2707. [Google Scholar] [CrossRef]
  150. Sandreschi, S.; Piras, A.M.; Batoni, G.; Chiellini, F. Perspectives on polymeric nanostructures for the therapeutic application of antimicrobial peptides. Nanomedicine 2016, 11, 1729–1744. [Google Scholar] [CrossRef] [PubMed]
  151. Riool, M.; de Breij, A.; Drijfhout, J.W.; Nibbering, P.H.; Zaat, S.A.J. Antimicrobial peptides in biomedical device manufacturing. Front. Chem. 2017, 5, 63. [Google Scholar] [CrossRef] [PubMed]
  152. Żelechowska, P.; Agier, J.; Brzezińska-Błaszczyk, E. Endogenous antimicrobial factors in the treatment of infectious diseases. Cent. Eur. J. Immunol. 2016, 41, 419–425. [Google Scholar] [CrossRef]
  153. Tasiemski, A.; Salzet, M.; Gaill, F.U.S. Antimicrobial Peptides. Patent No. 8,652,514, 2014. [Google Scholar]
  154. Deslouches, B.; Di, Y.P. Antimicrobial peptides with selective antitumor mechanisms: Prospect for anticancer applications. Oncotarget 2017, 8, 46635–46651. [Google Scholar] [CrossRef]
  155. Wang, G. Antimicrobial Peptides: Discovery, Design and Novel Therapeutic Strategies; CAB International: Wallingford, UK, 2017. [Google Scholar]
  156. 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]
  157. 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. [Google Scholar] [CrossRef]
  158. Nawrot, R.; Barylski, J.; Nowicki, G.; Broniarczyk, J.; Buchwald, W.; Goździcka-Józefiak, A.J.F. Plant antimicrobial peptides. Folia Microbiol. 2013, 59, 181–196. [Google Scholar] [CrossRef]
  159. 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]
  160. Espinosa-Leal, C.A.; Puente-Garza, C.A.; García-Lara, S.J.P. In vitro plant tissue culture means for production of biologically active compounds. Planta 2018, 248, 1–18. [Google Scholar] [CrossRef]
  161. Hussain, A.; Qarshi, I.A.; Nazir, H.; Ullah, I. Plant tissue culture: Current status and opportunities. In Recent Advances in Plant In Vitro Culture; Leva, A., Laura, M.R., Rinaldi, L.M.R., Eds.; Intech Open Limited: London, UK, 2012; pp. 1–28. [Google Scholar]
  162. Jiao, J.; Gai, Q.-Y.; Yao, L.-P.; Niu, L.-L.; Zang, Y.-P.; Fu, Y.-J. Ultraviolet radiation for flavonoid augmentation in Isatis tinctoria L. hairy root cultures mediated by oxidative stress and biosynthetic gene expression. Ind. Crops Prod. 2018, 118, 347–354. [Google Scholar] [CrossRef] [PubMed]
  163. Efferth, T. Biotechnology applications of plant callus cultures. Engineering 2019, 5, 50–59. [Google Scholar] [CrossRef]
  164. Zhang, L.; Rybczynski, J.; Langenberg, W.; Mitra, A.; French, R. An efficient wheat transformation procedure: Transformed calli with long-term morphogenic potential for plant regeneration. Plant Cell Rep. 2000, 19, 241–250. [Google Scholar] [CrossRef] [PubMed]
  165. Carciofi, M.; Blennow, A.; Nielsen, M.M.; Holm, P.B.; Hebelstrup, K.H. Barley callus: A model system for bioengineering of starch in cereals. Plant Methods 2012, 8, 36. [Google Scholar] [CrossRef] [PubMed]
  166. Imani, J.; Li, L.; Schaefer, P.; Kogel, K.H. STARTS–A stable root transformation system for rapid functional analyses of proteins of the monocot model plant barley. Plant J. 2011, 67, 726–735. [Google Scholar] [CrossRef] [PubMed]
  167. Gutierrez-Valdes, N.; Häkkinen, S.T.; Lemasson, C.; Guillet, M.; Oksman-Caldentey, K.-M.; Ritala, A.; Cardon, F. Hairy root cultures—A versatile tool with multiple applications. Front. Plant Sci. 2020, 11, 33. [Google Scholar] [CrossRef]
  168. Madeira, L.M.; Szeto, T.H.; Henquet, M.; Raven, N.; Runions, J.; Huddleston, J.; Garrard, I.; Drake, P.M.; Ma, J.K.C. High-yield production of a human monoclonal IgG by rhizosecretion in hydroponic tobacco cultures. Plant Biotechnol. J. 2016, 4, 615–624. [Google Scholar] [CrossRef] [PubMed]
  169. Drake, P.M.; Barbi, T.; Sexton, A.; McGowan, E.; Stadlmann, J.; Navarre, C.; Paul, M.J.; Ma, J.K.-C. Development of rhizosecretion as a production system for recombinant proteins from hydroponically cultivated tobacco. FASEB J. 2009, 23, 3581–3589. [Google Scholar] [CrossRef] [PubMed]
  170. Donini, M.; Lombardi, R.; Lonoce, C.; Di Carli, M.; Marusic, C.; Morea, V.; Di Micco, P. Antibody proteolysis: A common picture emerging from plants. Bioengineered 2015, 6, 299–302. [Google Scholar] [CrossRef]
  171. Lallemand, J.; Bouché, F.; Desiron, C.; Stautemas, J.; de Lemos Esteves, F.; Périlleux, C.; Tocquin, P. Extracellular peptidase hunting for improvement of protein production in plant cells and roots. Front. Plant Sci. 2015, 6, 37. [Google Scholar] [CrossRef] [PubMed]
  172. Lonoce, C.; Salem, R.; Marusic, C.; Jutras, P.V.; Scaloni, A.; Salzano, A.M.; Lucretti, S.; Steinkellner, H.; Benvenuto, E.; Donini, M. Production of a tumor-targeting antibody with a human-compatible glycosylation profile in N. benthamiana hairy root cultures. Biotechnol. J. 2016, 11, 1209–1220. [Google Scholar] [CrossRef]
  173. Holásková, E.; Galuszka, P.; Mičúchová, A.; Šebela, M.; Öz, M.T.; Frébort, I. Molecular farming in barley: Development of a novel production platform to produce human antimicrobial peptide LL-37. Biotechnol. J. 2018, 13, e1700628. [Google Scholar] [CrossRef]
  174. Ma, J.K.; Drake, P.M.; Christou, P. The production of recombinant pharmaceutical proteins in plants. Nat. Rev. Genet. 2003, 4, 94–805. [Google Scholar] [CrossRef]
  175. Makhzoum, A.; Benyammi, R.; Moustafa, K.; Trémouillaux-Guiller, J. Recent advances on host plants and expression cassettes’ structure and function in plant molecular pharming. BioDrugs 2014, 28, 145–159. [Google Scholar] [CrossRef] [PubMed]
  176. Iyappan, G.; Omosimua, R.O.; Sathishkumar, R. Enhanced production of therapeutic proteins in plants: Novel Expression Strategies. In Advances in Plant Transgenics: Methods and Applications; Springer: Berlin/Heidelberg, Germany, 2019; pp. 333–351. [Google Scholar]
  177. da Cunha, N.B.; Leite, M.L.; Dias, S.C.; Vianna, G.R.; Rech Filho, E.L. Plant genetic engineering: Basic concepts and strategies for boosting the accumulation of recombinant proteins in crops. IJLTES 2019, 8, 7–13. [Google Scholar]
  178. Zakharchenko, N.; Buryanov, Y.I.; Lebedeva, A.; Pigoleva, S.; Vetoshkina, D.; Loktyushov, E.; Chepurnova, M.; Kreslavski, V.; Kosobryukhov, A.J.R. Physiological features of rapeseed plants expressing the gene for an antimicrobial peptide cecropin P1. Russ. J. Plant Physiol. 2013, 60, 411–419. [Google Scholar] [CrossRef]
  179. Abdallah, N.A.; Shah, D.; Abbas, D.; Madkour, M. Stable integration and expression of a plant defensin in tomato confer resistance to fusarium wilt. GM Crop. 2010, 1, 344–350. [Google Scholar] [CrossRef]
  180. Ribeiro, T.P.; Arraes, F.B.M.; Lourenço-Tessutti, I.T.; Silva, M.S.; Lisei-de-Sá, M.E.; Lucena, W.A.; Macedo, L.L.P.; Lima, J.N.; Santos Amorim, R.M.; Artico, S.J.P. Transgenic cotton expressing Cry10Aa toxin confers high resistance to the cotton boll weevil. Plant Biotechnol. J. 2017, 15, 997–1009. [Google Scholar] [CrossRef]
  181. Ilyas, H.; Datta, A.; Bhunia, A. An approach towards structure-based antimicrobial peptide design for use in the development of transgenic plants: A strategy for plant disease management. Curr. Med. Chem. 2017, 24, 1350–1364. [Google Scholar] [CrossRef]
  182. 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]
  183. Sathoff, A.E.; Velivelli, S.; Shah, D.M.; Samac, D.A. Plant defensin peptides have antifungal and antibacterial activity against human and plant pathogens. Phytopathology 2019, 109, 402–408. [Google Scholar] [CrossRef]
  184. Lipsky, A.; Joshi, J.R.; Carmi, N.; Yedidia, I. Expression levels of antimicrobial peptide tachyplesin I in transgenic Ornithogalum lines affect the resistance to Pectobacterium infection. J. Biotechnol. 2016, 238, 22–29. [Google Scholar] [CrossRef]
  185. Weinhold, A.; Dorcheh, E.K.; Li, R.; Rameshkumar, N.; Baldwin, I.T. Antimicrobial peptide expression in a wild tobacco plant reveals the limits of host-microbe-manipulations in the field. eLife 2018, 7, e28715. [Google Scholar] [CrossRef] [PubMed]
  186. Varasteh-Shams, M.; Nazarian-Firouzabadi, F.; Ismaili, A. The direct and indirect transformation methods on expressing a recombinant Dermaseptin peptide in tobacco transgenic hairy root clones. Curr. Plant Biol. 2020, 24, 100177. [Google Scholar] [CrossRef]
  187. Li, F.; Shen, H.; Wang, M.; Fan, K.; Bibi, N.; Ni, M.; Yuan, S.; Wang, X. A synthetic antimicrobial peptide BTD-S expressed in Arabidopsis thaliana confers enhanced resistance to Verticillium dahliae. Mol. Genet. Genom. 2016, 291, 1647–1661. [Google Scholar] [CrossRef]
  188. Meng, D.-M.; Zhao, J.-F.; Ling, X.; Dai, H.-X.; Guo, Y.-J.; Gao, X.-F.; Dong, B.; Zhang, Z.-Q.; Meng, X.; Fan, Z.-C. Recombinant expression, purification, and antimicrobial activity of a novel antimicrobial peptide PaDef in Pichia pastoris. Protein Expr. Purif. 2017, 130, 90–99. [Google Scholar] [CrossRef] [PubMed]
  189. Chandrasekaran, J.; Brumin, M.; Wolf, D.; Leibman, D.; Klap, C.; Pearlsman, M.; Sherman, A.; Arazi, T.; Gal-On, A. Development of broad virus resistance in non-transgenic cucumber using CRISPR/Cas9 technology. Mol. Plant Pathol. 2016, 17, 1140–1153. [Google Scholar] [CrossRef] [PubMed]
  190. Abe, K.; Sakai, Y.; Nakashima, S.; Araki, M.; Yoshida, W.; Sode, K.; Ikebukuro, K. Design of riboregulators for control of cyanobacterial (Synechocystis) protein expression. Biotechnol. Lett. 2014, 36, 287–294. [Google Scholar] [CrossRef] [PubMed]
  191. 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]
  192. Yevtushenko, D.P.; Misra, S. Transgenic expression of antimicrobial peptides in plants: Strategies for enhanced disease resistance, improved productivity, and production of therapeutics. In Small Wonders: Peptides for Disease Control; Rajasekaran, K., Jeffrey, W.C., Jesse, M.J., Emilio, M., Eds.; ACS Publications: Washington, DC, USA, 2012; pp. 445–458. [Google Scholar]
  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] [PubMed]
  194. Bundo, 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]
  195. Fisher, A.C.; Kamga, M.-H.; Agarabi, C.; Brorson, K.; Lee, S.L.; Yoon, S. The current scientific and regulatory landscape in advancing integrated continuous biopharmaceutical manufacturing. Trends Biotechnol. 2019, 37, 253–267. [Google Scholar] [CrossRef] [PubMed]
  196. Streatfield, S.J. Approaches to achieve high-level heterologous protein production in plants. Plant Biotechnol. J. 2007, 5, 2–15. [Google Scholar] [CrossRef] [PubMed]
  197. Rage, E.; Marusic, C.; Lico, C.; Baschieri, S.; Donini, M. Current state-of-the-art in the use of plants for the production of recombinant vaccines against infectious bursal disease virus. Appl. Microbiol. Biotechnol. 2020, 104, 2287–2296. [Google Scholar] [CrossRef]
  198. Pogue, G.P.; Vojdani, F.; Palmer, K.E.; Hiatt, E.; Hume, S.; Phelps, J.; Long, L.; Bohorova, N.; Kim, D.; Pauly, M.; et al. Production of pharmaceutical-grade recombinant aprotinin and a monoclonal antibody product using plant-based transient expression systems. Plant Biotechnol. J. 2010, 8, 638–654. [Google Scholar] [CrossRef]
  199. Gleba, Y.; Klimyuk, V.; Marillonnet, S. Viral vectors for the expression of proteins in plants. Curr. Opin. Biotechnol. 2007, 18, 134–141. [Google Scholar] [CrossRef]
  200. Diamos, A.G.; Mason, H.S. High-level expression and enrichment of norovirus virus-like particles in plants using modified geminiviral vectors. Protein Expr. Purif. 2018, 151, 86–92. [Google Scholar] [CrossRef] [PubMed]
  201. Mbewana, S.; Mortimer, E.; Pêra, F.F.P.G.; Hitzeroth, I.I.; Rybicki, E.P. Production of H5N1 Influenza virus matrix protein 2 Ectodomain protein bodies in Tobacco plants and in insect cells as a candidate universal Influenza vaccine. Front. Bioeng. Biotechnol. 2015, 3, 197. [Google Scholar] [CrossRef] [PubMed]
  202. 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. 2013, 115, 99–106. [Google Scholar] [CrossRef]
  203. Herrera Diaz, A.; Kovacs, I.; Lindermayr, C. Inducible expression of the de-novo designed antimicrobial peptide SP1-1 in tomato confers resistance to Xanthomonas campestris pv. vesicatoria. PLoS ONE 2016, 11, e0164097. [Google Scholar] [CrossRef] [PubMed]
  204. Arya, S.S.; Rookes, J.E.; Cahill, D.M.; Lenka, S.K. Next-generation metabolic engineering approaches towards the development of plant cell suspension cultures as specialized metabolite producing biofactories. Biotechnol. Adv. 2020, 45, 107635. [Google Scholar] [CrossRef] [PubMed]
  205. 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 2019, 9, 30. [Google Scholar] [CrossRef]
  206. Rademacher, T.; Sack, M.; Blessing, D.; Fischer, R.; Holland, T.; Buyel, J. Plant cell packs: A scalable platform for recombinant protein production and metabolic engineering. Plant Biotechnol. J. 2019, 17, 1560–1566. [Google Scholar] [CrossRef]
  207. Poborilova, Z.; Plchova, H.; Cerovska, N.; Gunter, C.J.; Hitzeroth, I.I.; Rybicki, E.P.; Moravec, T. Transient protein expression in tobacco BY-2 plant cell packs using single and multi-cassette replicating vectors. Plant Cell Rep. 2020, 39, 1115–1127. [Google Scholar] [CrossRef]
  208. Gengenbach, B.B.; Opdensteinen, P.; Buyel, J.F. Robot cookies—Plant cell packs as an automated high-throughput screening platform based on transient expression. Front. Bioeng. Biotechnol. 2020, 8, 393. [Google Scholar] [CrossRef]
  209. Baker, D.; Sali, A. Protein structure prediction and structural genomics. Science 2001, 294, 93–96. [Google Scholar] [CrossRef] [PubMed]
  210. Bansal, A.K. Role of bioinformatics in the development of new antibacterial therapy. Expert Rev. Anti. Infect. Ther. 2008, 6, 51–65. [Google Scholar] [CrossRef] [PubMed]
  211. Franklin, T.J.; Snow, G.A. Biochemistry and Molecular Biology of Antimicrobial Drug Action, 5th ed.; Kluwer Academic: Norwell, MA, USA, 1998. [Google Scholar]
  212. Wang, G.; Li, X.; Wang, Z. APD3: The antimicrobial peptide database as a tool for research and education. Nucleic Acids Res. 2016, 44, D1087–D1093. [Google Scholar] [CrossRef] [PubMed]
  213. Chen, C.H.; Lu, T.K. Development and challenges of antimicrobial peptides for therapeutic applications. Antibiotics 2020, 9, 24. [Google Scholar] [CrossRef]
  214. Nguyen, L.T.; Chau, J.K.; Perry, N.A.; de Boer, L.; Zaat, S.A.; Vogel, H.J. Serum stabilities of short tryptophan and arginine-rich antimicrobial peptide analogs. PLoS ONE 2010, 5, e12684. [Google Scholar] [CrossRef]
  215. McPhee, J.B.; Hancock, R.E. Function and therapeutic potential of host defense peptides. J. Pept. Sci. 2005, 11, 677–687. [Google Scholar] [CrossRef]
  216. Starr, C.G.; Wimley, W.C. Antimicrobial peptides are degraded by the cytosolic proteases of human erythrocytes. Biochim. Biophys. Acta Biomembr. 2017, 1859, 2319–2326. [Google Scholar] [CrossRef]
  217. Brogden, N.K.; Brogden, K.A. Will new generations of modified antimicrobial peptides improve their potential as pharmaceuticals? Int. J. Antimicrob. Agents 2011, 38, 217–225. [Google Scholar] [CrossRef]
Figure 1. Representative examples of antimicrobial peptides produced in plant systems as effective antimicrobials (source: 3-dimensional structures downloaded from RCSB Protein Data Bank, https://www.rcsb.org/ (accessed on 20 August 2023).
Figure 1. Representative examples of antimicrobial peptides produced in plant systems as effective antimicrobials (source: 3-dimensional structures downloaded from RCSB Protein Data Bank, https://www.rcsb.org/ (accessed on 20 August 2023).
Life 13 01875 g001
Figure 2. Plant systems as ‘production platforms’ for molecular farming of novel and potent antimicrobial peptides.
Figure 2. Plant systems as ‘production platforms’ for molecular farming of novel and potent antimicrobial peptides.
Life 13 01875 g002
Figure 3. Significance of natural compounds and derivatives as potential antimicrobial agents.
Figure 3. Significance of natural compounds and derivatives as potential antimicrobial agents.
Life 13 01875 g003
Figure 4. Antimicrobial peptides: novel peptide-based therapeutics and their mechanisms.
Figure 4. Antimicrobial peptides: novel peptide-based therapeutics and their mechanisms.
Life 13 01875 g004
Table 1. Key examples of phytomolecules as potent antimicrobials, their bioactive constituents, and modes of action against pathogenic microbes (modified from Tiwari et al. [7]).
Table 1. Key examples of phytomolecules as potent antimicrobials, their bioactive constituents, and modes of action against pathogenic microbes (modified from Tiwari et al. [7]).
S. No.ClassificationKey ExamplesMechanism of ActionReference
1.Plant-derived compounds
PhenolicsThymol,
Carvacrol
The hydroxyl group increases the
Membrane disruption and
Leakage of cellular contents
[60]
Flavonoid compoundsCatechinsAntibacterial against Shigella, Vibrio, and Streptococcus mutans[74,75]
Hydroxylated phenolsCatechol and pyrogallolAntibacterial against Corynebacterium xerosis,
Pseudomonas putida, and
P. pyocyanea, and antifungal (catechol) against Penicillium italicum and Fusarium oxysporum
[76]
Polyphenol (3,5,4′-trihydroxystilbene)ResveratrolAntifungal against Plasmopara viticola, Sphaeropsis sapinea, and Pyricularia oryzae. In C. albicans, resveratrol penetrates the cell membrane and causes apoptosis.
Antibacterial against M. tuberculosis, VRE, S. typhimurium, and MRSA
[77,78,79,80]
Essential oil from Salvia fruticosa----Inhibition of Efflux pump in Staphylococcus epidermidis (clinical isolates)[81]
Quinones from Juglans and PlumbagoJuglone and plumbaginAntibacterial against S. aureus by increasing membrane permeability and restricting the formation of cell wall[82]
Essential oil from Chenopodium ambrosioides----Efflux pump Tet(K) inhibition in
S. aureus IS-58
[83]
AlkaloidCapsaicinEfflux pump NorA inhibition in
S. aureus SA-1199B
[84]
Anthraquinone
from Hypericum perforatum
HypericinAntimicrobial activity against methicillin-resistant and methicillin-sensitive Staphylococcus[85]
AlkaloidCatharanthineEfflux pump inhibition in
P. aeruginosa
[86]
Dimeric Phenylpropanoids from Styrax japonicaLignans
Styraxjaponoside C
Antifungal against C. albicans showing membrane-active mechanisms[87]
FlavonoidBaicaleinS. aureus SA-1199B NorA efflux pump inhibition[88]
TriterpenoidsUrsolic acid and derivativesinhibition of efflux pump AcrA/B, MacB, TolC and YojI in MDR E. coli (KG4)[89]
2.Plant by-products in food processing
Green husks of walnuts----Antibacterial against B. subtilis,
S. aureus and B. cereus
[90]
Grape pomacePhenolics Growth is hampered in S. aureus, yeasts, and Salmonella sp.[91]
Bergamot peel, an essential oil by-productChlorogenic acidAntibacterial against B. subtilis and food-borne E. coli, S. enterica[70]
Pomegranate
fruit peel extracts
Phenolic constituents Hampered growth in
S. aureus, Y. enterocolitica,
L. monocytogenes, etc.
[92]
Pomegranate rindTanninsAntimicrobial against L. monocytogenes modify microbial cell membranes and impair cell homeostasis[93]
Coconut huskTannins and Phenolic constituentsIn L. monocytogenes, and V. cholera, growth is hampered[72]
Olive juice powder and olive pomacePhenolic compound (oleocanthal)Antimicrobial against L. monocytogenes, S. aureus, and E. coli[94]
3. Animal-origin compounds
ChitosanPolycationic biopolymer
compound
Antibacterial towards
L. monocytogenes, B. cereus, S. aureus, and others
[64]
Milk-derived substances (casein and whey proteins)----Antibacterial/antifungal against Helicobacter, Listeria, Salmonella, Staphylococcus, E. coli, yeasts, and filamentous fungi[95]
LysozymeBacteriolytic enzymeLysozyme hydrolyzes
the β-1, 4 linkages between N-acetylmuramic acid and N-acetylglucosamine in the peptidoglycan of the microbial cell wall
[64]
4.Antimicrobials of bacterial origin
BacteriocinNisinGrowth is hampered in Gram-positive and spore-producing bacteria in food [96]
Reuterinβ-hydroxypropionaldehydeAntimicrobial towards foodborne pathogens[97]
5. Antimicrobials from algae and mushrooms
Phlorotannins from marine brown algae----Antimicrobial towards
S. aureus, Salmonella spp., etc.
[98]
Grifolin, and pleuromutilin from macrofungi----Antimicrobial activity against
S. aureus, B. cereus,
L. monocytogenes, E. coli
[99]
Fatty acids, β-carotene-linoleic acid, flavonoids from Agaricus spp.----Antimicrobial towards Micrococcus luteus,
B. cereus, etc.
[100]
Table 2. Representative examples of genome editing in biological organisms for applied biotechnologies.
Table 2. Representative examples of genome editing in biological organisms for applied biotechnologies.
S. No. Genome-
Editing Tool
Biological SystemMethod of Genome EditingApplicationsReference
1.Cas9/sgRNA
system
Cucumis
sativus
eIF4E (eukaryotic
translation initiation factor 4E) gene was targeted by Cas9/sgRNA construct
Plant resistance to viruses[189]
2.RNA interference (RNAi)Synechocystis sp.CRISPR-RNA (crRNA) in Synechocystis sp.
degrades target mRNA
To address drug resistance in
microbes
[190]
3.Genetic
engineering
Arabidopsis
thaliana
Cht1 signal peptide (Cht1SP)-
thanatin(S)-GFP construct was introduced in the plant
Antifungal and antibacterial activity of Thanatin(S), antimicrobial peptide[191]
4.Metabolic
engineering/
overexpression
Poplar spp.MsrA2 (antimicrobial peptide) overexpression in the plantImproved pathogen resistance[192]
5.De novo designing of AMP and engineering in plant Nicotiana
benthamiana
SPI-I (AMP) was designed and introduced in plant systemAntimicrobial function against pathogens[193]
6.Plant
transformation
Oryza sativaPlant transformation with cecropin A geneFungal and bacterial pathogen resistance[194]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tiwari, P.; Srivastava, Y.; Sharma, A.; Vinayagam, R. Antimicrobial Peptides: The Production of Novel Peptide-Based Therapeutics in Plant Systems. Life 2023, 13, 1875. https://doi.org/10.3390/life13091875

AMA Style

Tiwari P, Srivastava Y, Sharma A, Vinayagam R. Antimicrobial Peptides: The Production of Novel Peptide-Based Therapeutics in Plant Systems. Life. 2023; 13(9):1875. https://doi.org/10.3390/life13091875

Chicago/Turabian Style

Tiwari, Pragya, Yashdeep Srivastava, Abhishek Sharma, and Ramachandran Vinayagam. 2023. "Antimicrobial Peptides: The Production of Novel Peptide-Based Therapeutics in Plant Systems" Life 13, no. 9: 1875. https://doi.org/10.3390/life13091875

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop