Next Article in Journal
Effect of Daytime and Tree Canopy Height on Sampling of Cacopsylla melanoneura, a ‘Candidatus Phytoplasma mali’ Vector
Previous Article in Journal
The Active Compounds and Therapeutic Mechanisms of Pentaherbs Formula for Oral and Topical Treatment of Atopic Dermatitis Based on Network Pharmacology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Utility of Plant Growth-Promoting Rhizobacteria as Biological Control Agents towards Plant-Parasitic Nematodes

Department of Entomology and Plant Pathology, Auburn University, Auburn, AL 36849, USA
*
Authors to whom correspondence should be addressed.
P.S. and K.G. contributed equally to this work.
Plants 2020, 9(9), 1167; https://doi.org/10.3390/plants9091167
Submission received: 27 July 2020 / Revised: 28 August 2020 / Accepted: 2 September 2020 / Published: 9 September 2020

Abstract

:
Plant-parasitic nematodes (PPN) are among the most economically and ecologically damaging pests, causing severe losses of crop production worldwide. Chemical-based nematicides have been widely used, but these may have adverse effects on human health and the environment. Hence, biological control agents (BCAs) have become an alternative option for controlling PPN, since they are environmentally friendly and cost effective. Lately, a major effort has been made to evaluate the potential of a commercial grade strain of plant growth-promoting rhizobacteria (PGPR) as BCAs, because emerging evidence has shown that PGPR can reduce PPN in infested plants through direct and/or indirect antagonistic mechanisms. Direct antagonism occurs by predation, release of antinematicidal metabolites and semiochemicals, competition for nutrients, and niche exclusion. However, the results of direct antagonism may be inconsistent due to unknown endogenous and exogenous factors that may prevent PGPR from colonizing plant’s roots. On the other hand, indirect antagonism may occur from the induced systemic resistance (ISR) that primes whole plants to better fight against various biotic and abiotic constraints, actuating faster and/or stronger defense responses (adaption), enhancing their promise as BCAs. Hence, this review will briefly revisit (i) two modes of PGPR in managing PPN, and (ii) the current working models and many benefits of ISR, in the aim of reassessing current progresses and future directions for isolating more effective BCAs and/or developing better PPN management strategy.

Graphical Abstract

1. Introduction

Plant-parasitic nematodes (PPN) are biotrophic parasites that cause billions of dollars in economic damages on major food crops including corn (Zea mays L.), potato (Solanum tuberosum L.), soybean (Glycine max (L.) Merr.), as well as an important fiber crop, cotton (Gossypium hirsutum L.) [1]. Thus, a variety of management strategies using chemical nematicides, cultural control, resistant varieties and biological control (BCA) have long been tested, and employed to manage PPN. Nematicides, however, may not always be environmentally friendly, having hazardous and non-target impacts on flora and fauna including many natural enemies [2]. Although nematicides may initially reduce PPN populations and provide protection from their damage, these plants may allow the increase of populations at the end of the growing season. The following year will require an additional application of costly nematicides [3]. On the other hand, cultural control and resistant cultivars are not always available [4,5]. Hence, increasing attention has lately been drawn to the potential use of BCAs as an effective, inexpensive, and environmentally friendly tools of PPN management [3].
BCAs, a natural enemy or predator, can work by direct or indirect antagonism. Direct antagonism occurs in general through a predation, a release of toxic metabolites or a competition for nutrients and niche exclusion, whereas indirect antagonism can operate via induced systemic resistance (ISR) or systemic acquired resistance (SAR) [3]. ISR is defined as an enhanced plant resistance induced by plant growth-promoting rhizobacteria (PGPR) and typically employs jasmonic acid (JA) and ethylene (ET) hormone signaling [6]. SAR is also a form of induced plant resistance activated by an exposure to necrotizing biotrophs, which requires the accumulation of salicylic acid (SA) in infected and systemic tissues [7]. Over the last several decades, increasing efforts have been invested to examine the efficacy and practicality of PGPR in the hope of commercializing them as economic BCAs (Table 1, Table 2, Table 3 and Table 4). PGPR are well proven “effective biostimulants”, facilitating plant growth and productivity, and being concurrently able to improve plant resistance against a broad range of microbial pathogens and insect herbivores. Moreover, PGPR can prime plants to better adapt or acclimate towards various abiotic stresses including drought, salinity, and extreme temperatures [8]. Therefore, the successful development of commercial PGPR (BCAs) will greatly aid in upgrading plants’ own survival capacity against PPN as well as many other ecological constraints, without “trading off” yield potential. However, the current utility of PGPR in the field is still limited, largely due to our little knowledge of, in the molecular, biochemical and cellular levels, their mode of interactions with PPN and plants. Hence, this review will briefly revisit current working models of the mode of actions of PGPR with a focus of PPN management, and pinpoint information gaps within, which help revamp unique and alternative prospective for the future studies.

2. Recent Increases in Agronomic Burden by PPN

PPN, microscopic roundworms, belonging to the phylum Nematoda that are among the most abundant animal on earth; over 4100 species have been found in a variety of environmental conditions. The majority of PPN live in the film of moisture surrounding soil particles and plant roots. PPN have protrusible stylets or mouth spear to enter root tissues [1]. On the basis of their feeding habitats, PPN are chiefly classified into two groups, ectoparasites, and endoparasites. Ectoparasites feed from the outside of root surfaces without entering into plant roots. Thus, they could be more susceptible to environmental stresses and predators, and causing lesser damage to plant roots, than endoparasites. Endoparasites are able to penetrate completely or partly into plant roots during the infection process. This causes physical injury as well as allows secondary damages by bacteria and fungi infected along with or after PPN [113]. Damages caused by endoparasitic nematodes including Meloidogyne spp. (root-knot nematodes; RKN), Heterodera spp., Globodera spp. (cyst nematodes), Pratylenchus spp. (lesion nematodes), and Rotylenchulus spp. (reniform nematodes) are estimated to result in an annual loss of ~13% (~$216 billion) worldwide [114]. For instance, two genera of PPN, Meloidogyne spp. and Heterodera spp., alone cause the ~10% reduction of major food production including wheat (Triticum aestivum L.), rice (Oryza sativa L.), corn, potato, and sweet potato (Ipomoea batatas L.), which totaled an estimated loss of ~$80 to 114 billion per year. Rotylenchulus spp. has also emerged as a major threat in cash crops, cotton, and soybean, over the last decade throughout the southern regions of the US causing an estimated yield loss of over $200 million annually [1,115,116,117]. Hence, the establishment of effective and sustainable management program to control PPN has become an urgent task across the nations. However, current methods of integrated PPN management are rather restricted, when compared to that for other insect and animal pests, because PPN are microscopic and generally attack the underground portion of plants. The damage caused by PPN also are often not correctly identified because the aboveground symptoms mimic other diseases and environmental root-limiting factors delaying proper management [118,119].

Current Management Method against PPN

The current integrated pest management (IPM) program aimed at PPN largely strategizes the combined uses of crop rotation, tolerant/resistant cultivars, and nematicides. These methods, however, are still inadequate, expensive and/or cause numerous unexpected ecological and social drawbacks [120,121]. Presently, the most common strategy to manage PPN infections and field infestations is the application of nematicides (e.g., aldicarb, fluazaindolizine, fluensulfone, fluopyram, oxamyl, and terbufos) [122,123]. Nematicides have been an effective management tool; however, chemical nematicides are harmful to the environment, dangerous to apply and very costly [118]. Therefore, crop rotation has long been recommended as an alternative and potentially effective cultural practice to reduce nematode population density [5], but is not be a feasible option for all crops and farmers. For example, in the southern areas of the US, one of the most efficient crop rotations schemes for RKN, Meloidogyne incognita (Kafoid and White, 1919) [124] infested fields include the nonhost crop of peanut (Arachis hypogaea L.) [5,125]. However, peanuts require different harvesting equipment compared to cotton, soybeans, and corn, the most prominent row crops in the region. This harvesting equipment can range into the hundreds of thousands of dollars, and therefore may not be an economically feasible strategy.
Resistant cultivars increasingly have become available, preventing PPN from reproducing at as high a level as with susceptible cultivars; however, these cultivars are often accompanied by decreased yield [126]. Thus, some groups have advocated the use of tolerant cultivars that do not decrease the PPN reproduction but have little effect on yield potential [127]. The crucial drawback of tolerant cultivars though is that they cause an increase in the field population density of PPN. Thus, the use of resistant and tolerant cultivars is the most successful when used as a part of IPM system. Hence, alternative strategies must be looked at for effective, sustainable management of PPN. One, emerging option is biological control, which encompasses beneficial microbes such as PGPR that can reduce the impact of PPN, and other biotic attackers and abiotic stresses, as well as synergistically help the plant enhancing growth and development of various plant species [8].

3. Current utility of PGPR as BCAs of PPN; Mode of actions of PGPR

Recent studies have proposed that biological control could be a cost-effective approach to manage PPN when compared to the conventional methods. Especially, PGPR have emerged as promising BCA candidates [128,129]. PGPR are non-pathogenic bacteria, known to enhance plant growth and development in both non-stressed and stressed conditions by direct and indirect mechanisms [130]. The direct mechanism describes PGPR as bio-fertilizers producing organic compounds that promote plant growth or increase uptake of soil nutrients. Indirect mechanisms refer to PGPR-dependent biocontrol, including the production of antibiotics, Fe chelators (called as siderophores), and external cell wall degrading enzymes (e.g., chitinase and glucanase) that perhaps hydrolyze the pathogen (i.e., fungus) cell wall [131]. In line with this scenario, a number of PGPR, frequently from the genera of Agrobacterium, Bacillus, Paenibacillus and Pseudomonas, have been documented to reduce PPN population density through different mechanisms like parasitism, production of hydrolytic enzymes and antinematicidal metabolites (direct antagonism; Table 1), and inducing systemic resistance (indirect antagonism; Table 2) [130].

3.1. Direct Antagonism of PGPR against PPN

Direct antagonism of PGPR can prevent egg hatch, and/or the growth and reproduction of PPN, mainly through predation, and release of toxins or hydrolytic enzymes including hydrogen cyanide (HCN), 2,4-diacetylphloroglucinol (DAPG), chitinases, glucanases, proteases, and lipases (Table 1). The best characterized, although not necessarily PGPR, predatory bacterium is Pasteuria penetrans (ex Thorne, 1940) [42], which directly attacks and colonizes the body cavity of RKN (Meloidogyne spp.). Once attached on the cuticle surface of RKN, P. penetrans spores begin to form germination tubes that penetrate and rupture the cell membrane of PPN [132]. Likewise, P. thornei [133] can attack root-lesion nematodes (RLN, Pratylenchus spp.) and P. nishizawae [45] is able to attack the cysts nematode of two genera, Heterodera spp., and Globodera spp. [44].
Indeed, the exogenous application of P. penetrans demonstrated drastic decreases in the population density of M. javanica (Treub, 1885) [123] in sugarcane (Saccharum officinarum L.), by an average of 97.5% [43]. Thus, a few Pasteuria spp. have been made into commercial products; e.g., Clariva pn, Naviva ST and NewPro (Table 3). However, these products appear to be only active on one or two target species limiting actual practicality and use for field conditions. Producers often look for products that confer a broader spectrum of disease resistance, as well as defense responses against PPN.
On the other hand, a number of PGPR belonging to Bacillus spp. and Pseudomonas spp. produce nematicidal compounds. For example, several strains of Pseudomonas fluorescens (Flüge, 1886) [134] reduce potato cyst nematodes (PCN, Globodera spp.) and RKN (M. incognita) levels in vitro and/or in soil conditions by releasing DAPG, a phenolic antiphytopathogenic metabolite [49]. The wild type (WT) bacterium can reduce the motility of juvenile PCNs by 85%, comparing to 27% by mutant strains that disrupted DAPG productions [49]. On the other hand, P. fluorescens strain CHA0 and P. aeruginosa (Schröter, 1872) [135] produce the volatile compound HCN, an extremely poisonous toxin that adversely affects the proper functions and formations of mitochondria in serval RKN spp. (e.g., M. incognita and M. javanica) [46,50]. Interestingly, P. fluorescens strain CHA0 also releases both DAPG and pyoluteorin, two secondary metabolites that can significantly decrease population density of M. javanica in vitro and in a soil setting [51,53].
Several hydrolytic enzymes produced by PGPR are also able to inhibit the synthesis and maintenance of cell walls and membranes, as well as prevent the formation of cellular organelles [136]. Chitinases and discrete proteolytic enzymes produced from Bacillus cereus [137], B. firmus [138], B. licheniformis (Weigmann, 1898) [139], B. megaterium [140], and B. subtilis (Ehrenberg 1835) [141], as well as Corynebacterium paurometabolu [142] are proposed to be responsible for suppressing the multiplication of diverse RKN and cysts nematodes (see Table 1).
However, current attempts to commercialize PGPR for PPN management has had limited success [143]. A major concern has been that PGPR is inconsistent in managing PPN in the field, possibly due to many endogenous and exogenous factors limiting PGPR root colonization. Factors include other plants, variable soil conditions, and various rhizospheric metabolites and organisms [144]. Thus, there is a need to further investigate enhancing and/or optimizing the BCA activity of PGPR. Many studies now have turned into investigate the ability and a potential efficacy of indirect antagonism (i.e., ISR) of PGPR against PPN as an alternative, practical way to ensure the improvement of the defense capacity of plants against PPN, as well as other pathogens and abiotic constraints, and concurrently to enhance plant growth and development.

3.2. Indirect Antagonism, ISR of PGPR against PPN

Indirect antagonism of PGPR against PPN that occurs by ISR, is also referred to as PGPR-mediated priming, which systematically equips the "whole" plants to better cope with environmental constraints, actuating faster and/or stronger defense responses (adaption) to a subsequent exposure to various biotic and abiotic stresses. ISR is nonspecific in nature, and provides plants “a long-lasting protection” and “a broad-spectrum disease resistance (defense responses)” against various pathogenic microbes, insect herbivores and pests including PPNs; e.g., M. incognita, M. javanica, Heterodera glycines [145], H. cajani [146] and Globodera pallida [147] Behrens, 1975 (Table 2) [6]. In fact, ISR developed by various Bacillus spp. and Pseudomonas spp., including B. subtilis, B. amyloliquefaciens (ex Fukumoto, 1943) [148] and P. fluorescens have demonstrated promising results suppressing PPN in both dicotyledonous (Arabidopsis, bean (Fabaceae spp.), carnation (Dianthus caryophyllus L.), cucumber (Cucumis sativus L.), radish (Raphanus sativus L.), tobacco (Nicotiana tabacum L.), and tomato (Solanum lycopersicum L.)) as well as monocotyledonous (rice, maize, and sugarcane) plants (see Table 2).
However, molecular mechanisms underlying its occurrences are still elusive. Thus far, a majority of studies have foregrounded the potential, functional relevance of JA and ET hormones in ISR development, as mutant plants disrupted in JA/ET biosynthesis or signaling exhibited impaired PGPR-induced priming (reviewed in [6]). Moreover, exogenous application of JA could demonstrate ISR development against RKN (e.g., M. incognita, M. graminicola [149], and M javanica), cyst nematodes (e.g., H. avenae (Wollenweber, 1924) [150], H. schachtii [151]), and root lesion nematode (Pratylenchus neglectus (Rensch, 1924) [152]) in various plant species [153,154]. However, the root colonization of PGPR did not appear to stimulate the production of JA/ET in systemic (leaf) tissues, nor did they induce the expression of JA/ET-responsive genes in leaves [155,156]. Instead, ISR could potentiate JA/ET-responsive genes. When plants exposed/treated with PGPR were later challenged with other biotic stressors (e.g., microbial pathogens, insects, or herbivores), ISR facilitated a more rapid and/or enhanced level of induction of JA/ET-responsive genes, VSP, PDF1.2, and HEL [156,157].
On the other hand, selective results from earlier studies argued otherwise. Root inoculations of PGPR immediately triggered JA production and signaling transductions in systemic (leaf) tissues, which in turn heighted the state of defense responses throughout the plant [158]. Inoculation of PGPR Serrattia marcescens (Bizio, 1823) [159] strain 90–166 and P. fluorescens WCS417r showed rapid upregulation of several JA-biosynthetic and -responsive genes, e.g., VSP, PDF1.2, HEL, ChiB, LOX, and PAL in systemic (leaf) tissue throughout a number of crop plants, as well as Arabidopsis [61,160]. JA production then coordinates with abscisic acid (ABA) signaling to activate selective stress-responsive transcription factors such as OCP3 (Overexpressor of Cationic Peroxidase 3) and MYB60 [161,162], which in turn regulate stomatal closure, and suppress plant growth and yield [163]. These signaling and metabolic cascades may explain the inhibitory effect of JA in plant growth and development [164]. Indeed, the exogenous application of JA characteristically results in the inhibition of root growth [6,165,166], together suggesting that (i) ISR (and PGPR-induced JA signaling) could compete resource allocations (namely, defense and growth tradeoffs) with PGPR-mediated growth enhancement, or (ii) ISR employs alternative and/or additional (besides JA-responsive) signaling and/or metabolic pathways to prime plant disease resistance without reducing yield potential.

3.3. Potential Roles of SA in the Indirect Antagonism, SAR of PGPR against PPN

Recently, selective data argues that PGPR could stimulate SAR (instead of ISR), widely known to be developed by pathogenic microbes but not by PGPR [7,131]. The caveat is that ISR and SAR require mutually antagonistic cellular mechanisms, JA signaling for ISR versus SA signaling for SAR [6,7], which initially conflicted the role of SAR and SA signaling in the indirect antagonism of PGPR. However, a few studies reported that several Bacillus spp. can stimulate SA accumulations and signaling in plants (Table 2), hypothesizing that a mode of ISR mimics or shares SAR mechanisms. For instance, B. amyloliquefaciens MBI600, active ingredient of the biological fungicide Serifel®, was characterized to activate SA-dependent, but JA-independent, immunity, and reduce the disease severity of tomato spotted wilt virus in tomatoes [61]. It is notable that independent studies from other groups using similar PGPR strains such as B. amyloliquefaciens, B. mycoides (Flugge, 1886) [167], B. pumilus (Meyer and Gottheil, 1901) [168], and B. subtilis reported an induction of JA signaling (see Table 2). The cause for this contradiction is not understood, but may result from different experimental conditions and/or experimental errors. Alternatively, there may be timing differences to induce JA or SA signaling by PGPR (JA first and SA latter). A recent study using time-resolved transcription analyses indicated that the inoculation of Trichoderma harzianum (Rifai, 1969) [169] rapidly activates JA signaling on a time scale of hours, but later switches to enhance SA activity in 3 to 7 days in both infected and systemic tissues of plants [89]. This finding suggests the presence of complex and/or alternative mechanisms in PGPR-mediated systemic resistance. Further investigations must clarify the combined or antagonistic roles and functions of different plant hormone signaling such as JA, ET, SA, and ABA [170] in conveying defense activation by PGPR.

3.4. Crosstalk between SA and JA Signaling

As alluded, JA and SA signaling crosstalk with each other [171,172], and these interactions can be either antagonistic or synergistic. One of the first investigations into their interaction revealed an antagonistic relationship between JA and SA in tomatoes. They observed that aspirin, formulated SA, significantly suppressed the expression of JA marker genes [173]. Similarly, the expression of SA marker genes was inhibited upon the stimulation of JA by Pseudomonas syringae in tomato [174]. Furthermore, the application of exogenous JA also correlated with decreased SA activity, and enhanced JA signaling [175]. A more in depth look into the antagonism demonstrated that SA signaling targets the downstream of promoter in JA-responsive genes, and inhibits any further JA activity [176]. It is hypothesized that the antagonism be-tween the two hormones (JA and SA) helps plants conserve energy, and optimize the defense pathways in the presence of a single pathogen [171,172].
In contrast, a few early studies suggested a potential, synergistic relationship between JA and SA, as the pathogens Botrytis cinerea (Persoon, 1794) [177] and Tobacco mosaic virus stimulate both JA and SA at the same time [178,179]. Mur et al. later suggested that the concentration of hormones determined the interaction between them [180]. If the level of JA or SA is too high there was an antagonistic relationship; however, if the levels of each hormone were lower, the two hormones acted synergistically [180]. The exact concentration where synergism between the two hormones turns to antagonism is yet to be determined.
The goal of biological control research is to optimize the BCA to be as effective and efficient as possible against the target pathogen(s), and determining any manipulation of the JA and SA pathways by the target pathogen is an essential part of this type of research. This can, in turn, determine the best pathway to provide the most effective protection and defense against the target pathogen. This is important to keep in mind when selecting the best BCA in each management strategy.

4. Commercialization of PGPR

There is still a fundamental question (i.e., reproducibility) to address for the commercialization of PGPR, though multiple strains of PGPR are available on the market as biological nematicides (Table 3). However, the efficacy of these products needs to be further evaluated. To be commercially successful, PGPR products should have a broad application, long shelf life, be safe to use, have a viable market, easy availability, be consistent and have a low investment cost. Indeed, a large number of PGPR strains still remain to be investigated (Table 4). One particularly noticeable genus of PGPR is Bacillus spp. (9 direct antagonism, 7 indirect antagonism, and 17 uncharacterized); considered to be good options because they can quickly replicate and colonize plants, tolerate harsher environments, and easily form endospores. Moreover, they are documented to affect a broad spectrum of plant pathogens including PPN, viruses, bacteria, and fungi [181]. An extra benefit is that many of these Bacillus spp. can promote plant growth and help the plant adapt abiotic stresses, enhancing yield potential [182,183]. Bacillus spp. are the main species of commercialized PGPR due to their hardiness, as described above, compared to other effective PGPR such as Pseudomonas spp. [184]. More studies however are required to assess more effective mechanisms of their modus operandi as BCAs of PPN.

5. Conclusions

Emerging results have proposed the potential use and value of PGPR as BCAs in managing PPN. Selective strains of PGPR, in particular many Bacillus spp. appear to effectively suppress the growth and infestation of PPN in plants and fields, via direct or indirect antagonistic mechanism. Initially, much effort was invested to isolate (1) predatory PGPR strains and/or (2) PGPR-released nematicidal toxins and hydrolytic enzymes, which readily kill or inhibit PPN. However, their products were often highly host-specific, limiting actual use and practicality for field applications. Thus, recent studies have tried to understand, and improve the indirect antagonism (ISR) of PGPR, especially since PGPR have (i) longer shelf life, (ii) easy availability, (iii) low investment- and production-cost, and are (iv) safe to use. Moreover, PGPR are (v) effective biostimulants facilitating plant growth and yield potential, as well as are able to (vi) enhance plant resistance against a broad range of microbial pathogens, pests, and insect herbivores (i.e., ISR), and (vii) concurrently prime plants to better adapt or acclimate towards various abiotic stresses including drought, salinity, and extreme temperatures. These multifaceted activities, and benefits clearly underpin important merits and practicality for further screening and accessing the activity and efficacy of new PGPR in managing economically damaging PPN. Successful development/commercialization of BCAs will greatly aid in upgrading plants’ own survival capacity against various ecological constraints, but without trading off yield potential.

Author Contributions

Writing—original draft preparation, P.S., K.G., and S.-W.P.; writing—review and editing, W.L., K.S.L., and S.-W.P.; and funding acquisition, K.S.L. and S.-W.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported in part by the Alabama Cotton Commission, the Alabama Agricultural Experiment Station, and the Hatch program of the National Institute of Food and Agriculture (USDA).

Acknowledgments

The authors thank P. Donald and J. Coleman for critical comment on the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nicol, J.M.; Turner, S.J.; Coyne, D.L.; den Nijs, L.; Hockland, S.; Maafi, Z.T. Current nematode threats to world agriculture. In Genomics and Molecular Genetics of Plant-Nematode Interaction; Jones, J., Gheyse, G., Ffenoll, C., Eds.; Springer: Berlin/Heidelberg, Germany, 2011; pp. 21–43. [Google Scholar]
  2. Aktar, M.W.; Sengupta, D.; Chowdhury, A. Impact of pesticides use in agriculture: Their benefits and hazards. Interdiscip. Toxicol. 2009, 2, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Timper, P. Conserving and enhancing biological control of nematodes. J. Nematol. 2014, 46, 75–89. [Google Scholar] [PubMed]
  4. Xiang, N.; Lawrence, K.; Donald, P. Biological control potential of plant growth-promoting rhizobacteria suppression of Meloidogyne incognita on cotton and Heterodera glycines on soybean: A review. J. Phytopathol. 2018, 166, 449–458. [Google Scholar] [CrossRef] [Green Version]
  5. Kirkpatrick, T.L.; Sasser, J.N. Crop rotation and races of Meloidogyne incognita in cotton root-knot management. J. Nematol. 1984, 16, 323–328. [Google Scholar] [PubMed]
  6. Pieterse, C.M.J.; Zamioudis, C.; Berendsen, R.L.; Weller, D.M.; van Wees, S.C.M.; Bakker, P.A.H.M. Induced systemic resistance by beneficial microbes. Annu. Rev. Phytopathol. 2014, 52, 347–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Vlot, A.C.; Klessig, D.F.; Park, S.W. Systemic acquired resistance: The elusive signal(s). Curr. Opin. Plant Biol. 2008, 11, 436–442. [Google Scholar] [CrossRef] [Green Version]
  8. De Vrieze, J. The littlest farmhands. Science 2015, 349, 680–683. [Google Scholar] [CrossRef]
  9. Oka, Y.; Chet, I.; Spiegel, Y. Control of the rootknot nematode Meloidogyne javanica by Bacillus cereus. Biocontrol Sci. Technol. 1993, 3, 115–126. [Google Scholar] [CrossRef]
  10. Gao, H.; Qi, G.; Yin, R.; Zhang, H.; Li, C.; Zhao, X. Bacillus cereus strain S2 shows high nematicidal activity against Meloidogyne incognita by producing sphingosine. Sci. Rep. 2016, 6, 28756. [Google Scholar] [CrossRef]
  11. Ahmed, S. Bacillus cereus a potential strain infested cereal cyst nematode (Heterodera avenae). Pak. J. Nematol. 2019, 37, 53–61. [Google Scholar] [CrossRef]
  12. Ambo, P.B.N.; Ethiopia, E.A.; Serfoji, P.; Rajeshkumar, S.; Selvaraj, T. Management of root-knot nematode, Meloidogyne incognita on tomato cv Pusa Ruby by using vermicompost, AM fungus, Glomus aggregatum and mycorrhiza helper bacterium, Bacillus coagulans. J. Agric. Sci. Technol. 2010, 6, 37–45. [Google Scholar]
  13. Serfoji, P.; Smithra, P.; Saravavanan, K.; Durai Raj, K. Plant growth promotion and management of root-knot nematode Meloidogyne incognita through Glomus aggregatum and Bacillus coagulans and vermicomposting in tomato. Int. J. Pharm. Biol. Arch. 2013, 4, 532–536. [Google Scholar]
  14. Giannakou, I.O.; Karpouzas, D.G.; Prophetou-Athanasiadou, D. A novel non-chemical nematicide for the control of root-knot nematodes. Appl. Soil Ecol. 2004, 26, 69–79. [Google Scholar] [CrossRef]
  15. Mendoza, A.R.; Kiewnick, S.; Sikora, R.A. In vitro activity of Bacillus firmus against the burrowing nematode Radopholus similis, the root-knot nematode Meloidogyne incognita and the stem nematode Ditylenchus dipsaci. Biocontrol Sci. Technol. 2008, 18, 377–389. [Google Scholar] [CrossRef]
  16. Terefe, M.; Tefera, T.; Sakhuja, P.K. Effect of a formulation of Bacillus firmus on root-knot nematode Meloidogyne incognita infestation and the growth of tomato plants in the greenhouse and nursery. J. Invertebr. Pathol. 2009, 100, 94–99. [Google Scholar] [CrossRef]
  17. Terefe, M.; Tefera, T.; Sakhuja, P.K. Biocontrol (formulation of Bacillus firmus (BioNem)) of root-knot nematode, Meloidogyne incognita on tomato plants in the field. Ethiop. J. Agric. Sci. 2012, 22, 102–116. [Google Scholar]
  18. Xiong, J.; Zhou, Q.; Luo, H.; Xia, L.; Li, L.; Sun, M.; Yu, Z. Systemic nematicidal activity and biocontrol efficacy of Bacillus firmus against the root-knot nematode Meloidogyne incognita. World J. Microbiol. Biotechnol. 2015, 31, 661–667. [Google Scholar] [CrossRef]
  19. Geng, C.; Nie, X.; Tang, Z.; Zhang, Y.; Lin, J.; Sun, M.; Peng, D. A novel serine protease, Sep1, from Bacillus firmus DS-1 has nematicidal activity and degrades multiple intestinal-associated nematode proteins. Sci Rep. 2016, 6, 25012. [Google Scholar] [CrossRef] [Green Version]
  20. Bayer Crop Science. Available online: www.cropscience.bayer.us/products/seedgrowth/ponchovotivo/ (accessed on 18 August 2020).
  21. Siddiqui, Z.A.; Husain, S.I. Studies on the biological control of root-knot nematode. Curr. Nematol. 1991, 2, 5–6. [Google Scholar]
  22. Siddiqui, Z.A.; Mahmood, I. Biological control of root-rot disease complex of chickpea caused by Meloidogyne incognita Race 3 and Macrophomina phaseolina. Nematol. Medit. 1992, 20, 199–202. [Google Scholar]
  23. Jeong, M.-H.; Yang, S.-Y.; Lee, Y.-S.; Ahn, Y.-S.; Park, Y.-S.; Han, H.; Kim, K.-Y. Selection and characterization of Bacillus licheniformis MH48 for the biocontrol of pine wood nematode (Bursaphelenchus xylophilus). J. Korean For. Soc. 2015, 104, 512–518. [Google Scholar] [CrossRef] [Green Version]
  24. El-Nagdi, W.M.A.; Abd-El-Khair, H.; Soliman, G.M.; Ameen, H.H.; El-Sayed, G.M. Application of protoplast fusants of Bacillus licheniformis and Pseudomonas aeruginosa on Meloidogyne incognita in tomato and eggplant. Middle East J. Appl. Sci. 2019, 9, 622–629. [Google Scholar]
  25. Kloepper, J.W.; Beauchamp, C.J. A review of issues related to measuring of plant roots by bacteria. Can. J. Microbiol. 1992, 38, 1219–1232. [Google Scholar] [CrossRef]
  26. Padgham, J.L.; Sikora, R.A. Biological control potential and modes of action of Bacillus megaterium against Meloidogyne graminicola on rice. Crop Prot. 2007, 26, 971–977. [Google Scholar] [CrossRef]
  27. Mostafa, F.A.M.; Khalil, A.E.; Nour El-Deen, A.H.; Ibrahim, D.S. The role of Bacillus megaterium and other bio-agents in controlling root-knot nematodes infecting sugar beet under field conditions. Egypt. J. Biol. Pest Control 2018, 28, 66. [Google Scholar] [CrossRef]
  28. Lee, Y.S.; Kim, K.Y. Antagonistic potential of Bacillus pumilus L1 against root-knot nematode, Meloidogyne arenaria. J. Phytopathol. 2016, 164, 29–39. [Google Scholar] [CrossRef]
  29. Forghani, F.; Hajihassani, A. Recent advances in the development of environmentally benign treatments to control root-knot nematodes. Front. Plant Sci. 2020, 11, 1125. [Google Scholar] [CrossRef]
  30. Prakob, W.; Nguen-Hom, J.; Jaimasit, P.; Thanunchai, J.; Chaisuk, P. Biological control of lettuce root-knot disease by the used of Pseudomonas aeruginosa, Bacillus subtilis and Paecilomyces lilacinus. J. Agric. Technol. 2009, 13, 179–191. [Google Scholar]
  31. Kavitha, P.G.; Jonathan, E.L.; Nakkeeran, S. Effects of crude antibiotic of Bacillus subtilis on hatching of eggs and mortality of juveniles of Meloidogyne incognita. Nematol. Mediterr. 2012, 40, 211–215. [Google Scholar]
  32. Basyony, A.G.; Abo-Zaid, G.A. Biocontrol of the root-knot nematode, Meloidogyne incognita, using an eco-friendly formulation from Bacillus subtilis, lab and greenhouse studies. Egypt. J. Biol. Pest Control 2018, 28, 87. [Google Scholar] [CrossRef]
  33. de Mazzuchelli, R.C.L.; Mazzuchelli, E.H.L.; de Araujo, F.F. Efficiency of Bacillus subtilis for root-knot and lesion nematodes management in sugarcane. Biol. Control 2020, 143, 104185. [Google Scholar] [CrossRef]
  34. Gautam, A.; Siddiqui, Z.; Mahmood, I. Integrated management of Meloidogyne incognita on tomato. Nematol. Mediterr. 1995, 23, 245–248. [Google Scholar]
  35. Noel, G.R. Evaluation of thuringiensin for control of Heterodera glycines on soybean. J. Nematol. 1990, 22, 763–766. [Google Scholar] [PubMed]
  36. Wei, J.-Z.; Hale, K.; Carta, L.; Platzer, E.; Wong, C.; Fang, S.-C.; Aroian, R.V. Bacillus thuringiensis crystal proteins that target nematodes. Proc. Natl. Acad. Sci. USA 2003, 100, 2760–2765. [Google Scholar] [CrossRef] [Green Version]
  37. Mohammed, S.; Saedy, M.; Enan, M.; Ibrahim, N.E.; Ghareeb, A.; Moustafa, S. Biocontrol efficiency of Bacillus thuringiensis toxins against root-knot nematode, Meloidogyne incognita. J. Cell Mol. Biol. 2008, 7, 57–66. [Google Scholar]
  38. Mena, J.; Pimentel, E. Mechanism of action of Corynebacterium pauronetabolum strain C-924 on nematodes. Nematology 2002, 4, 287. [Google Scholar]
  39. Mankau, R.; Imbriani, J.L.; Bell, A.H. SEM observations on nematode cuticle penetration by Bacillus penetrans. J. Nematol. 1976, 8, 179–181. [Google Scholar]
  40. Mankau, R.; Prasad, N. Possibilities and problems in use of a sporozoan endoparasite for biological control of plant parasitic nematodes. Nematropica 1977, 2, 7–8. [Google Scholar]
  41. Dube, B.; Smart, G.C. Biological control of Meloidogyne incognita by Paecilomyces lilacinus and Pasteuria penetrans. J. Nematol. 1987, 19, 222–227. [Google Scholar]
  42. Sayre, R.M.; Starr, M.P. Pasteuria penetrans a mycelial and endospore-forming bacterium parasitic in plant-parasitic nematodes. Proc. Helminthol. Soc. Wash 1985, 52, 149–165. [Google Scholar]
  43. Bhuiyan, S.A.; Garlick, K.; Anderson, J.M.; Wickramasinghe, P.; Stirling, G.R. Biological control of root-knot nematode on sugarcane in soil naturally or artificially infested with Pasteuria penetrans. Australas. Plant Pathol. 2018, 47, 45–52. [Google Scholar] [CrossRef]
  44. Atibalentja, N.; Noel, G.R.; Domier, L.L. Phylogenetic position of the North American isolate of Pasteuria that parasitizes the soybean cyst nematode, Heterodera glycines, as inferred from 16S RDNA sequence analy-sis. Int. J. Syst. Evol. Microbiol. 2000, 50, 605–613. [Google Scholar] [CrossRef] [PubMed]
  45. Sayre, R.M.; Wergin, W.P.; Schmldt, J.M.; Starr, M.P. Pasteuria nishizawae sp. nov., a mycelial and endospore-forming bacterium parasitic on cyst nematodes of genera Heterodera and Globodera. Res. Microbiol. 1991, 142, 551–564. [Google Scholar] [CrossRef] [Green Version]
  46. Siddiqui, I.A.; Ehteshamul-Haque, S. Suppression of the root rot–root knot disease complex by Pseudomo-nas aeruginosa in tomato: The influence of inoculum density, nematode populations, moisture and other plant-associated bacteria. Plant Soil 2001, 237, 81–89. [Google Scholar] [CrossRef]
  47. Gallagher, L.A.; Manoil, C. Pseudomonas aeruginosa PAO1 kills Caenorhabditis elegans by cyanide poisoning. J. Bacteriol. 2001, 183, 6207–6214. [Google Scholar] [CrossRef] [Green Version]
  48. Singh, P.; Siddiqui, Z.A. Biocontrol of root-knot nematode Meloidogyne incognita by the isolates of Pseudomonas on tomato. Arch. Phytopathol. Plant Protect. 2010, 43, 1423–1434. [Google Scholar] [CrossRef]
  49. Cronin, D.; Moenne-Loccoz, Y.; Fenton, A.; Dunne, C.; Dowling, D.N.; O’gara, F. Role of 2,4-diacetyl-phloroglucinol in the interactions of the biocontrol Pseudomonad strain F113 with the potato cyst nematode Globodera rostochiensis. Appl. Environ. Microbiol. 1997, 63, 1357–1361. [Google Scholar] [CrossRef] [Green Version]
  50. Siddiqui, I.; Shaukat, S. Suppression of root-knot disease by Pseudomonas fluorescens CHA0 in tomato: Importance of bacterial secondary metabolite, 2,4-diacetylpholoroglucinol. Soil Biol. Biochem. 2003, 35, 1615–1623. [Google Scholar] [CrossRef]
  51. Hamid, M.; Siddiqui, I.A.; Shahid Shaukat, S. Improvement of Pseudomonas fluorescens CHA0 biocontrol activity against root-knot nematode by the addition of ammonium molybdate. Lett. Appl. Microbiol. 2003, 36, 239–244. [Google Scholar] [CrossRef]
  52. Siddiqui, I.A.; Haas, D.; Heeb, S. Extracellular protease of Pseudomonas fluorescens CHA0, a biocontrol factor with activity against the root-knot nematode Meloidogyne incognita. Appl. Environ. Microbiol. 2005, 71, 5646–5649. [Google Scholar] [CrossRef] [Green Version]
  53. Timper, P.; Koné, D.; Yin, J.; Ji, P.; McSpadden Gardener, B.B. Evaluation of an antibiotic-producing strain of Pseudomonas fluorescens for suppression of plant-parasitic nematodes. J. Nematol. 2009, 41, 234–240. [Google Scholar]
  54. Khan, M.R.; Mohidin, F.A.; Khan, U.; Ahamad, F. Native Pseudomonas spp. suppressed the root-knot nematode in in vitro and in vivo, and promoted the nodulation and grain yield in the field grown mungbean. Biol. Control 2016, 101, 159–168. [Google Scholar] [CrossRef]
  55. Zabaketa-Mejia, E. The effect of soil bacteria on Meloidogyne incognita (Kofoid & White) Chitwood infection. Diss. Abstr. Interact. 1985, 46, 1018. [Google Scholar]
  56. Rahul, S.; Chandrashekhar, P.; Hemant, B.; Chandrakant, N.; Laxmikant, S.; Satish, P. Nematicidal activity of microbial pigment from Serratia marcescens. Nat. Prod. Res. 2014, 28, 1399–1404. [Google Scholar] [CrossRef] [PubMed]
  57. Hasky-Guenther, K.; Hoffmann-Hergarten, S.; Sikora, R.A. Resistance against the potato cyst nematode Globodera pallida systemically induced by the rhizobacteria Agrobacterium radiobacter (G12) and Bacillus sphaericus (B43). Fundam. Appl. Nematol. 1998, 21, 511–517. [Google Scholar]
  58. Hackenberg, C.; Sikora, R.A. Influence of cultivar, temperature, and soil moisture on the antagonistic potential of Agrobacterium radiobacter against Globodera pallida. J. Nematol. 1992, 24, 594. [Google Scholar]
  59. Racke, J.; Sikora, R.A. Influence of the plant health-promoting rhizobacteria Agrobacterium radiobacter and Bacillus sphaericus on Globodera pallida root infection of potato and subsequent plant growth. J. Phytopathol. 1992, 134, 198–208. [Google Scholar] [CrossRef]
  60. Hackenberg, C.; Vrain, T.C.; Sikora, R.A. Rhizosphere colonization pattern of Agrobacterium Radiobacter strain G12A, an antagonistic rhizobacterium to the potato cyst nematode Globodera pallida. Microbiol. Res. 1999, 154, 57–61. [Google Scholar] [CrossRef]
  61. Ryu, C.-M.; Farag, M.A.; Hu, C.-H.; Reddy, M.S.; Kloepper, J.W.; Paré, P.W. Bacterial volatiles induce systemic resistance in Arabidopsis. Plant Physiol. 2004, 134, 1017–1026. [Google Scholar] [CrossRef] [Green Version]
  62. Beris, D.; Theologidis, I.; Skandalis, N.; Vassilakos, N. Bacillus amyloliquefaciens strain MBI600 induces Sali-cylic acid dependent resistance in tomato plants against tomato spotted wilt virus and potato virus Y. Sci. Rep. 2018, 8, 10320. [Google Scholar] [CrossRef] [Green Version]
  63. Burkett-Cadena, M.; Kokalis-Burelle, N.; Lawrence, K.S.; van Santen, E.; Kloepper, J.W. Suppressiveness of root-knot nematodes mediated by rhizobacteria. Biol. Control. 2008, 47, 55–59. [Google Scholar] [CrossRef]
  64. Choudhary, D.K.; Johri, B.N. Interactions of Bacillus spp. and plants—with special reference to induced systemic resistance (ISR). Microbiol. Res. 2009, 164, 493–513. [Google Scholar] [CrossRef]
  65. Li, J.; Zou, C.; Xu, J.; Ji, X.; Niu, X.; Yang, J.; Huang, X.; Zhang, K.-Q. Molecular mechanisms of nematode-nematophagous microbe interactions: Basis for biological control of plant-parasitic nematodes. Annu. Rev. Phytopathol. 2015, 53, 67–95. [Google Scholar] [CrossRef]
  66. Xiang, N.; Lawrence, K.S.; Kloepper, J.W.; Donald, P.A.; McInroy, J.A.; Lawrence, G.W. Biological control of Meloidogyne incognita by spore-forming plant growth-promoting rhizobacteria on cotton. Plant Dis. 2016, 101, 774–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Xie, S.; Jiang, H.; Ding, T.; Xu, Q.; Chai, W.; Cheng, B. Bacillus amyloliquefaciens FZB42 represses plant MiR846 to induce systemic resistance via a jasmonic acid-dependent signalling pathway. Mol. Plant Pathol. 2018, 19, 1612–1623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. de Halfeld-Vieira, B.A.; Vieira Júnior, J.R.; da Romeiro, R.S.; Silva, H.S.A.; Baracat-Pereira, M.C. Induction of systemic resistance in tomato by the autochthonous phylloplane resident Bacillus cereus. Pesqui. Agropecu. Bras. 2006, 41, 1247–1252. [Google Scholar] [CrossRef] [Green Version]
  69. Niu, D.-D.; Liu, H.-X.; Jiang, C.-H.; Wang, Y.-P.; Wang, Q.-Y.; Jin, H.-L.; Guo, J.-H. The plant growth-promoting rhizobacterium Bacillus cereus AR156 induces systemic resistance in Arabidopsis thaliana by simultaneously activating salicylate- and jasmonate/ethylene-dependent signaling pathways. Mol. Plant Microbe Interact. 2011, 24, 533–542. [Google Scholar] [CrossRef] [Green Version]
  70. Jiang, C.; Fan, Z.; Li, Z.; Niu, D.; Li, Y.; Zheng, M.; Wang, Q.; Jin, H.; Guo, J. Bacillus cereus AR156 triggers induced systemic resistance against Pseudomonas syringae pv. tomato DC3000 by suppressing MiR472 and activating CNLs-mediated basal immunity in Arabidopsis. Mol. Plant Pathol. 2020, 21, 854–870. [Google Scholar] [CrossRef]
  71. Liu, K.; Garrett, C.; Fadamiro, H.; Kloepper, J.W. Induction of systemic resistance in chinese cabbage against black rot by plant growth-promoting rhizobacteria. Biol. Control. 2016, 99, 8–13. [Google Scholar] [CrossRef] [Green Version]
  72. Bargabus, R.L.; Zidack, N.; Sherwood, J.E.; Jacobsen, B.J. Characterisation of systemic resistance in sugar beet elicited by a non-pathogenic, phyllosphere-colonizing Bacillus mycoides biological control agent. Physiol. Mol. Plant Pathol. 2002, 61, 289–298. [Google Scholar] [CrossRef]
  73. Ryu, C.-M.; Farag, M.A.; Hu, C.-H.; Reddy, M.S.; Wei, H.-X.; Paré, P.W.; Kloepper, J.W. Bacterial volatiles promote growth in Arabidopsis. Proc. Natl. Acad. Sci. USA 2003, 100, 4927–4932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhang, S.; Moyne, A.-L.; Reddy, M.S.; Kloepper, J.W. The role of salicylic acid in induced systemic resistance elicited by plant growth-promoting rhizobacteria against blue mold of tobacco. Biol. Control. 2002, 25, 288–296. [Google Scholar] [CrossRef]
  75. Bargabus, R.; Zidack, N.; Sherwood, J.; Jacobsen, B. Screening for the identification of potential biological control agents that induce systemic acquired resistance in sugar beet. Biol. Control Biol. Control 2004, 30, 342–350. [Google Scholar] [CrossRef]
  76. Kavitha, J.; Jonathan, E.I.; Umamaheswari, R. Field application of Pseudomonas fluorescens, Bacillus subtilis and Trichoderma viride for the control of Meloidogyne incognita (Kofoid and White) chitwood on sugarbeet. J. Biol. Control 2007, 21, 211–215. [Google Scholar]
  77. Choudhary, D.K.; Prakash, A.; Johri, B.N. Induced systemic resistance (ISR) in plants: Mechanism of action. Indian J. Microbiol. 2007, 47, 289–297. [Google Scholar] [CrossRef] [Green Version]
  78. Lastochkina, O.; Pusenkova, L.; Yuldashev, R.; Babaev, M.; Garipova, S.; Blagova, D.; Khairullin, R.; Aliniaeifard, S. Effects of Bacillus subtilis on some physiological and biochemical parameters of Triticum aestivum L. (wheat) under salinity. Plant Physiol. Biochem. 2017, 121, 80–88. [Google Scholar] [CrossRef]
  79. Akram, W.; Mahboob, A.; Javed, A.A. Bacillus thuringiensis strain 199 can induce systemic resistance in tomato against Fusarium wilt. Eur. J. Microbiol. Immunol. 2013, 3, 275–280. [Google Scholar] [CrossRef] [Green Version]
  80. Zuckerman, B.M.; Dicklow, M.B.; Acosta, N. A strain of Bacillus thuringiensis for the control of plant-parasitic nematodes. Biocontrol Sci. Technol. 1993, 3, 41–46. [Google Scholar] [CrossRef]
  81. Audenaert, K.; Pattery, T.; Cornelis, P.; Höfte, M. Induction of systemic resistance to Botrytis cinerea in tomato by Pseudomonas aeruginosa 7NSK2: Role of salicylic acid, pyochelin, and pyocyanin. Mol. Plant Microbe Interact. 2002, 15, 1147–1156. [Google Scholar] [CrossRef] [Green Version]
  82. Fatima, S.; Anjum, T. Identification of a potential ISR determinant from Pseudomonas aeruginosa PM12 against Fusarium wilt in tomato. Front. Plant Sci. 2017, 8, 848. [Google Scholar] [CrossRef]
  83. Krechel, A.; Faupel, A.; Hallmann, J.; Ulrich, A.; Berg, G. Potato-associated bacteria and their antagonistic potential towards plant-pathogenic fungi and the plant-parasitic nematode Meloidogyne incognita (Kofoid & White) Chitwood. Can. J. Microbiol. 2002, 48, 772–786. [Google Scholar] [PubMed]
  84. Saikia, S.K.; Tiwari, S.; Pandey, R. Rhizospheric biological weapons for growth enhancement and Meloidogyne incognita management in Withania somnifera cv. Poshita. Biol. Control. 2013, 65, 225–234. [Google Scholar] [CrossRef]
  85. De Vleesschauwer, D.; Djavaheri, M.; Bakker, P.A.H.M.; Höfte, M. Pseudomonas fluorescens WCS374r-induced systemic resistance in rice against Magnaporthe oryzae is based on pseudobactin-mediated priming for a salicylic acid-repressible multifaceted defense response. Plant Physiol. 2008, 148, 1996–2012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Leeman, M.; van Pelt, J.A.; den Ouden, F.M.; Heinsbroek, M.; Bakker, P.A.H.M.; Schippers, B. Induction of systemic resistance by Pseudomonas fluorescens in radish cultivars differing in susceptibility to Fusarium wilt, using a novel bioassay. Eur. J. Plant Pathol. 1995, 101, 655–664. [Google Scholar] [CrossRef]
  87. Almaghrabi, O.A.; Massoud, S.I.; Abdelmoneim, T.S. Influence of inoculation with plant growth promoting rhizobacteria (PGPR) on tomato plant growth and nematode reproduction under greenhouse conditions. Saudi J. Biol. Sci. 2013, 20, 57–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Reitz, M.; Rudolph, K.; Schröder, I.; Hoffmann-Hergarten, S.; Hallmann, J.; Sikora, R.A. lipopolysaccharides of Rhizobium etli strain G12 act in potato roots as an inducing agent of systemic resistance to infection by the cyst nematode Globodera pallida. Appl. Environ. Microbiol. 2000, 66, 3515–3518. [Google Scholar] [CrossRef] [Green Version]
  89. Martínez-Medina, A.; Fernandez, I.; Lok, G.B.; Pozo, M.J.; Pieterse, C.M.J.; Wees, S.C.M.V. Shifting from priming of salicylic acid- to jasmonic acid-regulated defences by Trichoderma protects tomato against the root knot nematode Meloidogyne incognita. New Phytol. 2017, 213, 1363–1377. [Google Scholar] [CrossRef] [Green Version]
  90. BioNemaGonTM. Available online: http://www.agrilife.in/biopesti_microrigin_nemagon.htm (accessed on 18 August 2020).
  91. Clariva® pn. Available online: https://www.syngenta-us.com/seed-treatment/clariva-pn (accessed on 18 August 2020).
  92. Nematode: Alternative Controls. Available online: www.attra.ncat.org/attra-pub/PDF/nematode.pdf (accessed on 18 August 2020).
  93. MeloCon®WG. Available online: https://www.certisusa.com/products/bionematicides/melocon-wg (accessed on 18 August 2020).
  94. Mistures Comprising a Bacillus Strain and a Pesticide. Available online: https://patentswarm.com/patents/US10251400B2 (accessed on 18 August 2020).
  95. US EPA, Pesticide Product Label, NewPro. Available online: https://www3.epa.gov/pesticides/chem_search/ppls/085004-00011-20130422.pdf (accessed on 18 August 2020).
  96. Nortica 10 WP. Available online: http://www.tomirwin.com/pdf/labels/Nortica%2010WP.pdf (accessed on 18 August 2020).
  97. VoTIVo FS. Available online: https://agrobaseapp.com/united-states/pesticide/votivo-fs (accessed on 18 August 2020).
  98. Bansal, R.K.; Verma, V. kumar. Antagonistic efficacy of Azotobacter chroococcum against Meloidogyne javanica infecting brinjal. Indian J. Nematol. 2002, 32, 132–134. [Google Scholar]
  99. Crow, W.T. Effects of a commercial formulation of Bacillus firmus I-1582 on golf course bermudagrass infested with Belonolaimus longicaudatus. J. Nematol. 2014, 46, 331–335. [Google Scholar]
  100. Tiwari, S.; Pandey, S.; Chauhan, P.; Pandey, R. Biocontrol agents in co-inoculation manages root knot nematode [Meloidogyne incognita (Kofoid & White) Chitwood] and enhances essential oil content in Ocimum basilicum L. Ind. Crops Prod. 2017, 97, 292–301. [Google Scholar]
  101. Siddiqui, Z.A.; Shakeel, U. Screening of Bacillus isolates for potential biocontrol of the wilt disease complex of pigeon pea (Cajanus cajan) under greenhouse and small-scale field conditions. J. Plant Pathol. 2007, 89, 179–183. [Google Scholar]
  102. Zhou, L.; Yuen, G.; Wang, Y.; Wei, L.; Ji, G. Evaluation of bacterial biological control agents for control of rootknot nematode disease on tomato. Crop Prot. 2016, 84, 8–13. [Google Scholar] [CrossRef]
  103. Khan, M.R.; Akram, M. Effect of certain antagonistic fungi and rhizobacteria on wilt disease complex caused by Meloidogyne incognita and Fusarium oxysporium f. sp. lycopersici on tomato. Nematol. Mediterr. 2000, 28, 139–144. [Google Scholar]
  104. Meyer, S.L.F.; Massood, S.I.; Chitwood, D.J.; Roberts, D.P. Evaluation of Trichoderma virens and Burkholderia cepacia for antagonistic activity against root-knot nematode, Meloidogyne incognita. Nematology 2000, 2, 871–879. [Google Scholar]
  105. Colagiero, M.; Rosso, L.C.; Ciancio, A. Diversity and biocontrol potential of bacterial consortia associated to root-knot nematodes. Biol. Control. 2018, 120, 11–16. [Google Scholar] [CrossRef]
  106. Son, S.H.; Khan, Z.; Kim, S.G.; Kim, Y.H. Plant growth-promoting rhizobacteria, Paenibacillus polymyxa and Paenibacillus lentimorbus suppress disease complex caused by root-knot nematode and Fusarium wilt fungus. J. Appl. Microbiol. 2009, 107, 524–532. [Google Scholar] [CrossRef] [PubMed]
  107. Oliveira, D.F.; Campos, V.P.; Amaral, D.R.; Nunes, A.S.; Pantaleão, J.A.; Costa, D.A. Selection of rhizobacteria able to produce metabolites active against Meloidogyne exigua. Eur. J. Plant Pathol. 2007, 119, 477–479. [Google Scholar] [CrossRef]
  108. Kermarrec, A.; Jacqua, G.; Anais, J. Effect of Fusarium solani and Pseudomonas solanacearum on the infestation of aubergine with the plant-parasitic nematode Rotylenchulus reniformis. Nematologica 1994, 40, 152–154. [Google Scholar]
  109. Siddiqui, Z.A.; Singh, L.P. Effects of fly ash, Pseudomonas striata and Rhizobium on the reproduction of nematode Meloidogyne incognita and on the growth and transpiration of pea. J. Environ. Biol. 2005, 26, 117–122. [Google Scholar]
  110. Seenivasan, N.; Parameswaran, S.; Sridar, P.; Gopalakrishnan, C.; Gnanamurthy, P. Application of Bioagents and Neem Cake as Soil Application for the Management of Root-knot Nematode in Turmeric. National Congress on Centenary of Nematology in India Appraisal and Future Plans; Indian Agricultural Research Institute: New Delhi, India, 2001; p. 164. [Google Scholar]
  111. Insunza, V.; Alström, S.; Eriksson, K.B. Root bacteria from nematicidal plants and their biocontrol potential against trichodorid nematodes in potato. Plant Soil. 2002, 241, 271–278. [Google Scholar] [CrossRef]
  112. Rashad, F.; Fathy, H.; Samir, A.; Elghonaimy, A. Isolation and characterization of multifunctional Streptomyces species with antimicrobial, nematicidal and phytohormone activities from marine environments in Egypt. Microbiol. Res. 2015, 175, 34–47. [Google Scholar] [CrossRef] [PubMed]
  113. Hussey, R.S.; Grundler, F.M. Nematode parasitism of plants. In Physiology and Biochemistry of Free-Living and Plant Parasitic Nematodes; Perry, R.N., Wright, J., Eds.; 11 CAB International Press: Oxford, UK, 1988; pp. 213–243. [Google Scholar]
  114. Singh, S.; Singh, B.; Singh, A.P. Nematodes: A threat to sustainability of agriculture. Procedia Environ. Sci. 2015, 29, 215–216. [Google Scholar] [CrossRef] [Green Version]
  115. Sikkens, R.B.; Weaver, D.B.; Lawrence, K.S.; Moore, S.R.; van Santen, E. LONREN upland cotton germ-plasm response to Rotylenchulus reniformis inoculum level. Nematropica 2011, 6, 68–74. [Google Scholar]
  116. Roshi, R.A.; King, R.L.; Lawrence, G.W. Classification of Rothylenhulus reniformis numbers in cotton using remotely sensed hyperspectral data on self-organizing maps. J. Nematol. 2010, 42, 179–193. [Google Scholar]
  117. Weaver, D.B. Cotton nematodes. In Cotton, 2nd ed.; Fang, D.D., Rercy, R.G., Eds.; ASA, CSSA, and SSSA: Madison, WI, USA, 2015; pp. 547–570. [Google Scholar]
  118. Elhady, A.; Giné, A.; Topalovic, O.; Jacquiod, S.; Sørensen, S.J.; Sorribas, F.J.; Heuer, H.; Castagnone-Sereno, P. Microbiomes associated with infective stages of root-knot and lesion nematodes in soil. PLoS ONE 2017, 12, e0177145. [Google Scholar] [CrossRef] [Green Version]
  119. Bernard, G.C.; Egnin, M.; Bonsi, C. The impact of plant-parasitic nematodes on agriculture and methods of control. In Nematology—Concepts, Diagnosis and Control; Shah, M.M., Mohammod, M., Eds.; IntechOpen: London, UK, 2017; pp. 121–151. [Google Scholar]
  120. Pimentel, D. Environmental and economic costs of the application of pesticides primarily in the United States. Environ. Dev. Sustain. 2005, 7, 229–252. [Google Scholar] [CrossRef]
  121. Mitiku, M. Plant-parasitic nematodes and their management: A review. Agric. Res. Technol. Open Access J. 2016, 16, 555980. [Google Scholar] [CrossRef]
  122. Shepherd, R.L.; Huck, M.G. Progression of root-knot nematode symptoms and infection on resistant and susceptible cottons. J. Nematol. 1989, 2, 235–241. [Google Scholar]
  123. Rhoads, M.L. Cholinesterase in the parasitic nematode, Stephanurus denatus. J. Biochem. 1981, 17, 9316–9323. [Google Scholar]
  124. Chitwood, B.G. “Root-knot Nematodes”—Part I. A revision of the genus Meloidogyne goeldi, 1887. Proc. Helminthol. Soc. Wah. 1949, 16, 90–104. [Google Scholar]
  125. Johnson, A.; Dowler, C.; Handoo, Z. Population dynamics of Meloidogyne incognita, M. arenaria and other nematodes and crop yields in rotations of cotton, peanut, and wheat under minimum tillage. J. Nematol. 2000, 32, 52–61. [Google Scholar] [PubMed]
  126. Vyska, M.; Cunniffe, N.; Gilligan, C. Trade-off between disease resistance and crop yield: A landscape-scale mathematical modeling perspective. J. R. Soc. Interface 2016, 13, 20160451. [Google Scholar] [CrossRef] [Green Version]
  127. Trudgill, D.L. Resistance to and tolerance of plant parasitic nematodes in plants. Ann. Rev. Phytopathol. 1991, 29, 167–192. [Google Scholar] [CrossRef]
  128. Siddiqui, Z.A.; Mahmood, I. Role of bacteria in the management of plant parasitic nematodes: A Review. Bioresour. Technol. 1999, 69, 167–179. [Google Scholar] [CrossRef]
  129. Wani, A.H. Plant growth-promoting rhizobacteria as biocontrol agents of phytonematodes. In Biocontrol Agents of Phytonematodes; Askary, T.H., Martinelli, P.R.P., Eds.; CAB International: Wallingford, UK, 2015; pp. 339–362. [Google Scholar]
  130. Lugtenberg, B.; Kamilova, F. Plant-growth-promoting rhizobacteria. Annu. Rev. Microbiol. 2009, 63, 541–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Van Loon, L.C.; Bakker, P.A.H.M.; Pieterse, C.M.J. Systemic resistance induced by rhizosphere bacteria. Annu. Rev. Phytopathol. 1998, 36, 453–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Bishop, A. Pasteuria penetrans and its parasitic interaction with plant parasitic nematodes. In Endospore-Forming Soil Bacteria; Logan, N.A., de Vos, P., Eds.; Springer: Berlin/Heidelberg, Germany, 2011; pp. 181–201. [Google Scholar]
  133. Starr, M.P.; Sayre, R.M. Pasteuria thornei sp. nov. and Pasteuria penetrans sensu stricto emend., mycelial and endospore-forming bacteria parasitic, respectively, on plant-parasitic nematodes of the genera Pratylenchus and Meloidogyne. Ann. Inst. Pasteur. Microbiol. 1988, 139, 11–31. [Google Scholar] [CrossRef]
  134. Migula, W. Über ein neues System der Bakterien. Arab Bacteriol. Inst. Karlsr. 1984, 1, 235–328. [Google Scholar]
  135. Migula, W. System der Bakteriem; Gustav Fischer: Jena, Germany, 1900; Volume 2. [Google Scholar]
  136. Maksimov, I.V.; Abizgil’dina, R.R.; Pusenkova, L.I. Plant growth promoting rhizobacteria as alternative to chemical crop protectors from pathogens (Review). Appl. Biochem. Microbiol. 2011, 47, 333–345. [Google Scholar] [CrossRef]
  137. Frankland, S.G.; Frankland, R.F. Studies on some new microorganisms obtained from air. Royal Soc. Lon-don Phil. Trans. Ser. B Biol. Sci. 1887, 178, 257–287. [Google Scholar]
  138. Bredemann, G.; Werner, W. Botanische beschreibung haufinger am buttersaureabbau beteiligter sporenbildender bakterienspezies. Zentralbl. Bakteriol. Parasitenkd. Infektionskr. Hyg. Abt. II 1933, 87, 446–475. [Google Scholar]
  139. Chester, F.D. A Manual of Determinative Bacteriology; The Macmillan Co.: New York, NY, USA, 1901; pp. 1–401. [Google Scholar]
  140. De Bary, A. Vergleichende Morphologie und Biologie der Pilze, Mycetozoen und Bacterien; Wilhelm Engelmann: Leipzig, Germany, 1884. [Google Scholar]
  141. Cohn, F. Untersuchungen uber Bakterien. Beitr. Biol. Pflanz. 1872, 1, 127–224. [Google Scholar]
  142. Collins, M.D.; Smida, J.; Dorsch, M.; Stackebrandt, E. Tsukamurella gen. nov. harboring Corynebacterium paurometabolum and Rhodococcus aurantiacus. Int. J. Syst. Bacteriol. 1988, 38, 385–391. [Google Scholar] [CrossRef]
  143. Ramamoorthy, V.; Viswanathan, R.; Raguchander, T.; Prakasam, V.; Samiyappan, R. Induction of systemic resistance by plant growth promoting rhizobacteria in crop plants against pests and diseases. Crop Prot. 2001, 20, 1–11. [Google Scholar] [CrossRef]
  144. Babalola, O.O. Beneficial bacteria of agricultural importance. Biotechnol. Lett. 2010, 32, 1559–1570. [Google Scholar] [CrossRef]
  145. Ichinohe, M. On the soybean nematode, Heterodera glycines n. sp., from Japan (trans.). Oyo-Dobutsugaku-Zasshi 1952, 17, 1–4. [Google Scholar]
  146. Koshy, P.K. A new species of Heterodera from India. Indian Phytopathol. 1967, 20, 272–274. [Google Scholar]
  147. Stone, A.R. Heterodera pallida n. sp. (Nematoda: Heteroderidae), a second species of potato cyst nematode. Nematologica 1973, 18, 591–606. [Google Scholar] [CrossRef]
  148. Priest, F.G.; Goodfellow, M.; Shute, L.A.; Berkeley, R.C.W. Bacillus amyloliquefaciens sp. nov., nom. rev. Int. J. Syst. Bacteriol. 1987, 37, 69–71. [Google Scholar] [CrossRef]
  149. Golden, A.M.; Birchfield, W. Rice root-knot nematode (Meloidogyne graminicola) as a new pest of rice. Plant Dis. Rep. 1968, 52, 423. [Google Scholar]
  150. Krall, E.L.; Krall, H.A. Revision of the plant nematodes of the family Heteroderidae on the basis of the trophic specialization of these parasites and their co-evolution with their host plants. In Fitogel’Mintologi-Cheskie Issledovaniya. Moscow; Nauka: Moscow, USSR, 1978; pp. 39–56. [Google Scholar]
  151. Schmidt, A. Über den Rüben-Nematoden (Heterodera schachtii A.S.). Z. Ver. Die Rüben-Zucker-Ind. Zollverein 1871, 21, 1–19. [Google Scholar]
  152. Filipjev, I.N.; Schuurmans Stekhoven, J.H., Jr. A Manual of Agricultural Helminthology; Brill: Leiden, The Netherland, 1941; p. 878. [Google Scholar]
  153. Nahar, K.; Kyndt, T.; De Vleesschauwer, D.; Höfte, M.; Gheysen, G. The jasmonate pathway is a key player in systemically induced defense against root knot nematodes in rice. Plant Physiol. 2011, 157, 305–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Fujimoto, T.; Tomitaka, Y.; Abe, H.; Tsuda, S.; Futai, K.; Mizukubo, T. Expression profile of jasmonic acid-induced genes and the induced resistance against the root-knot nematode (Meloidogyne incognita) in tomato plants (Solanum lycopersicum ) after foliar treatment with methyl jasmonate. J. Plant Physiol. 2011, 168, 1084–1097. [Google Scholar] [CrossRef] [PubMed]
  155. Hubbard, K.E.; Nishimura, N.; Hitomi, K.; Getzoff, E.D.; Schroeder, J.I. Early abscisic acid signal transduction mechanisms: Newly discovered components and newly emerging questions. Genes Dev. 2010, 24, 1695–1708. [Google Scholar] [CrossRef] [Green Version]
  156. Lee, S.C.; Luan, S. ABA signal transduction at the crossroad of biotic and abiotic stress responses. Plant Cell Environ. 2012, 35, 53–60. [Google Scholar] [CrossRef]
  157. Daszkowska-Golec, A.; Szarejko, I. The molecular basis of ABA-mediated plant response to drought. In Abiotic Stress—Plant Responses and Applications in Agriculture; Vahdati, K., Leslie, C., Eds.; IntechOpen: Rijeka, Croatia, 2013; Available online: https://www.intechopen.com/books/abiotic-stress-plant-responses-and-applications-in-agriculture/the-molecular-basis-of-aba-mediated-plant-response-to-drought (accessed on 3 September 2020).
  158. Der Ent, S.V.; Hulten, M.V.; Pozo, M.J.; Czechowski, T.; Udvardi, M.K.; Pieterse, C.M.J.; Ton, J. Priming of plant innate immunity by rhizobacteria and β-aminobutyric acid: Differences and similarities in regulation. New Phytol. 2009, 183, 419–431. [Google Scholar] [CrossRef] [Green Version]
  159. de Toni, J.B.; Trevisan, V. Schizomycetaceae Naeg. In Sylloge Fungorum Omnium Hujusque Cognitorum; Saccardo, P.A., Ed.; Sumptibus Auctoris: Berlin, Germany, 1889; Volume 8, pp. 923–1087. [Google Scholar]
  160. Pieterse, C.M.; van Wees, S.C.; van Pelt, J.A.; Knoester, M.; Laan, R.; Gerrits, H.; Weisbeek, P.J.; van Loon, L.C. A novel signaling pathway controlling induced systemic resistance in Arabidopsis. Plant Cell 1998, 10, 1571–1580. [Google Scholar] [CrossRef] [Green Version]
  161. Ramírez, V.; Van der Ent, S.; García-Andrade, J.; Coego, A.; Pieterse, C.M.; Vera, P. OCP3 is an important modulator of NPR1-mediated jasmonic acid-dependent induced defenses in Arabidopsis. BMC Plant Biol. 2010, 10, 199. [Google Scholar] [CrossRef] [Green Version]
  162. Cominelli, E.; Galbiati, M.; Vavasseur, A.; Conti, L.; Sala, T.; Vuylsteke, M.; Leonhardt, N.; Dellaporta, S.L.; Tonelli, C. A guard-cell-specific MYB transcription factor regulates stomatal movements and plant drought tolerance. Curr. Biol. 2005, 15, 1196–1200. [Google Scholar]
  163. Lim, C.W.; Baek, W.; Jung, J.; Kim, J.-H.; Lee, S.C. Function of ABA in stomatal defense against biotic and drought stresses. Int. J. Mol. Sci. 2015, 16, 15251–15270. [Google Scholar] [CrossRef] [Green Version]
  164. Huang, H.; Gao, H.; Liu, B.; Qi, T.; Tong, J.; Xiao, L.; Xie, D.; Song, S. Arabidopsis MYB24 regulates jasmonate-mediated stamen development. Front. Plant Sci. 2017, 8, 1525. [Google Scholar] [CrossRef]
  165. Feys, B.; Benedetti, C.E.; Penfold, C.N.; Turner, J.G. Arabidopsis mutants selected for resistance to the phytotoxin coronatine are male sterile, insensitive to methyl jasmonate, and resistant to a bacterial pathogen. Plant Cell 1994, 6, 751–759. [Google Scholar] [CrossRef] [Green Version]
  166. Mosblech, A.; Thurow, C.; Gatz, C.; Feussner, I.; Heilmann, I. Jasmonic acid perception by COI1 involves inositol polyphosphates in Arabidopsis thaliana. Plant J. 2011, 65, 949–957. [Google Scholar] [CrossRef]
  167. Liu, Y.; Lai, Q.; Shao, Z. Genome analysis-based reclassification of Bacillus weihenstephanensis as a later heterotypic synonym of Bacillus mycoides. Int. J. Syst. Evol. Microbiol. 2018, 68, 106–112. [Google Scholar] [CrossRef]
  168. Meyer, A.; Gottheil, O. Botanische beschreibung einiger bodenbakterien. Zentralbl. Bakteriol. Parasitenkd. Infektionskr. Hyg. Abt. II 1901, 7, 680–691. [Google Scholar]
  169. Chaverri, P.; Samuels, G.J. Hypocrea lixii, the teleomorph of Trichoderma harzianum. Mycol. Prog. 2002, 1, 283–286. [Google Scholar] [CrossRef]
  170. Martinez- Medina, A.; Appels, F.V.W.; van Wees, S.C.M. Impact of salicylic acid- and jasmonic acid-defences on root colonization of Trichoderma harzianum T-78. Plant Signal. Behav. 2017, 12, 1345414. [Google Scholar] [CrossRef] [Green Version]
  171. Glazebrook, J. Contrasting mechanisms of defense against biotrophic and necrotrophic pathogens. Ann. Rev. Phytopathol. 2005, 43, 205–227. [Google Scholar] [CrossRef]
  172. Gimenez-Ibanez, S.; Solano, R. Nuclear jasmonate and salicylate signaling and crosstalk in defense against pathogens. Front. Plant Sci. 2013, 4, 72. [Google Scholar] [CrossRef] [Green Version]
  173. Pena-Cortés, H.; Albrecht, T.; Prat, S.; Weiler, E.W.; Willmitzer, L. Aspirin prevents wound-induced gene expression in tomato leaves by blocking jasmonic acid biosynthesis. Planta 1993, 191, 123–128. [Google Scholar] [CrossRef]
  174. Uppalapati, S.R.; Ishiga, Y.; Wangdi, T.; Kunkel, B.N.; Anand, A.; Mysore, K.S.; Bender, C.L. The phytotoxin coronatine contributes to pathogen fitness and is required for suppression of salicylic acid accumulation in tomato inoculated with Pseudomonas syringae pv. tomato DC3000. Mol. Plant Microbe Interact. 2007, 20, 955–965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Tamaoki, D.; Seo, S.; Yamada, S.; Kano, A.; Miyamoto, A.; Shishido, H.; Miyoshi, S.; Taniguchi, S.; Akimitsu, K.; Gomi, K. Jasmonic acid and salicylic acid activate a common defense system in rice. Plant Signal. Behav. 2013, 8, 24260. [Google Scholar] [CrossRef] [PubMed]
  176. Van der Does, D.; Leon-Reyes, A.; Koornneef, A.; Van Verk, M.C.; Rodenburg, N.; Pauwels, L.; Goossens, A.; Körbes, A.P.; Memelink, J.; Ritsema, T.; et al. Salicylic acid suppresses jasmonic acid signaling downstream of SCFCOI1-JAZ by targeting GCC promoter motifs via transcription factor ORA59. Plant Cell. 2013, 25, 744–761. [Google Scholar] [CrossRef] [Green Version]
  177. Whetzel, H.H. A synopsis of the genera and species of the Sclerotiniaceae, a family of stromatic inoperculate discomycetes. Mycologia 1945, 37, 648–714. [Google Scholar] [CrossRef]
  178. Mitter, N.; Kazan, K.; Way, H.M.; Broekaert, W.F.; Manners, J.M. Systemic induction of an Arabidopsis plant defensin gene promoter by tobacco mosaic virus and jasmonic acid in transgenic tobacco. Plant Sci. 1998, 136, 169–180. [Google Scholar] [CrossRef]
  179. Clarke, J.D.; Volko, S.M.; Ledford, H.; Ausubel, F.M.; Dong, X. Roles of salicylic acid, jasmonic acid, and ethylene in cpr-induced resistance in Arabidopsis. Plant Cell 2000, 12, 2175–2190. [Google Scholar] [CrossRef] [Green Version]
  180. Mur, L.A.J.; Kenton, P.; Atzorn, R.; Miersch, O.; Wasternack, C. The outcomes of concentration-specific interactions between salicylate and jasmonate signaling include synergy, antagonism, and oxidative stress leading to cell death. Plant Physiol. 2006, 140, 249–262. [Google Scholar] [CrossRef] [Green Version]
  181. Shafi, J.; Tian, H.; Ji, M. Bacillus species as versatile weapons for plant pathogens: A review. Biotechnol. Biotechnol. Equip. 2017, 31, 446–459. [Google Scholar] [CrossRef] [Green Version]
  182. Kloepper, J.W.; Lifshitz, R.; Zablotowicz, R. Free-living bacterial inocula for enhancing crop productivity. Trends Biotechnol. 1989, 7, 39–44. [Google Scholar] [CrossRef]
  183. Vacheron, J.; Desbrosses, G.; Bouffaud, M.-L.; Touraine, B.; Moënne-Loccoz, Y.; Muller, D.; Legendre, L.; Wisniewski-Dyé, F.; Prigent-Combaret, C. Plant growth-promoting rhizobacteria and root system functioning. Front. Plant Sci. 2013, 4, 356. [Google Scholar] [CrossRef] [Green Version]
  184. Kloepper, J.W.; Ryu, C.-M.; Zhang, S. Induced systemic resistance and promotion of plant growth by Bacillus spp. Phytopathology 2004, 94, 1259–1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. A list of plant growth-promoting rhizobacteria (PGPR) conferring direct antagonisms against plant-parasitic nematodes (PPN).
Table 1. A list of plant growth-promoting rhizobacteria (PGPR) conferring direct antagonisms against plant-parasitic nematodes (PPN).
PGPR Target PPNMolecules or ModesReferences
Bacillus cereusHeterodera avenae,
Meloidogyne incognita,
Meloidogyne javanica
Sphingosine, Protease, Chitinase, Antibiotic production, Secondary metabolitesOka et al., 2008 [9]; Gao et al., 2016 [10]; Ahmed, 2019 [11]
Bacillus coagulansMeloidogyne incognitaHydrolytic enzymesAmbo et al., 2010 [12]; Serfoji et al., 2013 [13]; Xiang et al., 2018 [4]
Bacillus firmusDitylenchus dipasi, Heterodera spp., Meloidogyne incognita, Pratylenchus spp., Radopholus similis Sep 1 protease, Secondary metabolitesGiannakou et al., 2004 [14]; Mendoza et al., 2008 [15]; Terefe et al., 2009 [16]; Terefe et al., 2012 [17]; Xiong et al., 2015 [18]; Geng et al., 2016 [19]; Bayer Crop Science [20]
Bacillus licheniformisBursaphelenchulus xylophi lus, Meloidogyne incognitaProtease, ChitinaseSiddiqui and Husain 1991 [21]; Siddiqui and Mahmood 1992 [22]; Jeong et al., 2015 [23]; El-Nagdi et al., 2019 [24]
Bacillus megateriumHeterodera glycines,Meloidogyne incognita,
Meloidogyne graminicola
Protease, Secondary metabolitesKloepper et al., 1992 [25]; Padgham et al., 2007 [26]; Mostafa et al., 2018 [27]
Bacillus pumilus L1Heterodera glycines,
Meloidogyne arenaria
Protease, ChitinaseLee and Kim 2015 [28]; Forghani and Hajihassani et al., 2020 [29]
Bacillus subtilisHelicotylenchus multicinctus,
Meloidogyne graminicola, Meloidogyne incognita, Meloidogyne javanica, Rotylenchulus reniformis
Lipopeptide antibiotics, Hydrolytic enzymes, Secondary metabolitesPrakob et al., 2009 [30]; Kavitha et al., 2012 [31]; Basyony and Abo-Zaid, 2018 [32]; Mazzuchelli et al., 2020 [33]; Gautam et al., 1995 [34]
Bacillus thuringiensisHeterodera glycines, Meloidogyne incognitaBt crystal protein (toxin protein), Thuringiensin (β-exotoxin)Noel, 1990 [35]; Wei et al., 2003 [36]; Mohammed et al., 2008 [37]
Corynebacterium paurometabolumMeloidogyne incognitaHydrogen sulfide, ChitinaseMena and Pimentel, 2002 [38]
Pasteuria penetrans1Meloidogyne spp.PredationMankau et al., 1976 [39]; Mankau and Prasad, 1977 [40]; Dube and Smart, [41]; Sayre and Starr 1975 [42]; Bhuiyan et al., 2018 [43]
Pasteuria thornei1Pratylenchus spp. PredationMankau et al., 1976 [41]; Atibalentja et al., 2000 [44]
Pasteuria nishizawae1Globodera spp.,
Heterodera spp.
PredationSayre and Wergin, 1991 [45]
Pseudomonas aeruginosaCaenorhabditis elegans, Meloidogyne incognita, Meloidogyne javanicaHydrogen cyanide (HCN)Siddiqui and Ehteshamul-Haque, 2001 [46]; Gallagher and Manoil, 2001 [47]; Singh and Siddiqui, 2010 [48]
Pseudomonas. fluorescens F113Globodera rostochinensis2,4-diacetylphloroglucinol (DAPG)Cronin et al., 1997 [49]
Pseudomonas fluorescens CHA0Meloidogyne incognita, MeloidogynejavanicaHCN, DAPG, Pyoluteorin, Extracellular proteaseSiddiqui and Shaukat, 2003 [50]; Hamid et al., 2003 [51]; Siddiqui et al., 2005 [52]
Pseudomonas fluorescens Wood1RMeloidogyne incognitaDAPGTimper et al., 2009 [53]
Pseudomonas stutzeriMeloidogyne incognitaHCNKhan et al., 2016 [54]
Serratia marcescensMeloidogyne incognita,
Meloidogyne javanica,
Radopholus similis
Volatile metabolites, ProdigiosinZabaketa-Mejia, 1985 [55]; Rahul et al., 2014 [56]
1 nematode parasitic bacteria.
Table 2. A list of PGPR conferring indirect antagonisms against PPN.
Table 2. A list of PGPR conferring indirect antagonisms against PPN.
PGPRTarget PPNModesReferences
Agrobacterium radiobacter (G12)Globodera spp.ISRHasky-Guenther et al., 1998 [57]; Hackenberg and Sikora, 1992 [58]; Racke and Sikora, 1992 [59]; Hackenberg et al., 1999 [60]
Bacillus amyloliquefaciens
(syn. Bacillus velezensis)
Heterodera glycine,
Meloidogyne incognita
ISR and SARRyu et al., 2004 [61]; Beris et al., 2018 [62]; Burkett- Cadena et al., 2008 [63]; Choudhary et al., 2009 [64]; Li et al., 2015 [65]; Xiang et al., 2016 [66]; Xie et al., 2018 [67]
Bacillus cereusMeloidogyne javanica, Meloidogyne incognitaISRXiang et al., 2016 [66]; Halfeld-Vieira et al., 2006 [68]; Niu et al., 2011 [69]; Jiang et al., 2020 [70]
Bacillus mojavensisMeloidogyne incognitaISRXiang et al., 2016 [66]; Liu et al., 2016 [71]
Bacillus mycoidesMeloidogyne incognitaISR and SARXiang et al., 2016 [66]; Barbagus et al., 2004 [72]
Bacillus pasteuriiMeloidogyne incognitaISRXiang et al., 2016 [66]; Ryu et al., 2003 [73]
Bacillus pumilusHeterodera glycine,
Meloidogyne incognita
ISR and SARXiang et al., 2016 [66]; Zhang et al., 2002 [74]; Barbagus et al., 2004 [75]; Kavitha et al., 2007 [76]; Choudhary et al., 2007 [77]; Lastochkina et al., 2017 [78]
Bacillus sphaericusGlobodera pallida, Meloidogyne incognitaISRHasky-Guenther et al., 1998 [57]; Racke and Sikora 1992 [59]; Xiang et al., 2016 [66]
Bacillus subtilisHeterodera cajani, Meloidogyne arenaria, Meloidogyne incognita, Meloidogyne javanicaISR and SARRyu et al., 2004 [61]; Xiang et al., 2016 [66]; Kavitha et al., 2007 [76]; Choudhary et al., 2007 [77]; Lastochkina et al., 2017 [78]
Bacillus thuringiensisAphelenchus avenae, Meloidogyne incognitaISRZhang et al., 2002 [74] Akram et al., 2013 [79]; Zuckerman et al.; 1993 [80]
Pseudomonas aeruginosaMeloidogyne javanicaISR and SARAudenaert et al.,2013 [81]; Fatima et al., 2017 [82]
Pseudomonas fluorescensMeloidogyne incognita, Meloidogyne javanicaISR and SARSiddiqui and Shaukat 2003 [50]; Choudhary et al., 2007 [77]; Krechel et al., 2002 [83]; Saikia et al., 2013 [84]; de Vleesschauwer et al., 2012 [85]; Leeman et al., 1995 [86]
Pseudomonas putidaMeloidogyne incognitaISRKrechel et al., 2002 [83]; Almaghrabi et al., 2013 [87]
Rhizobium etliMeloidogyne spp.ISRReitz et al., 2000 [88]
Serratia marcescensMeloidogyne incognitaISRZhang et al., 2002 [74]; Almaghrabi et al., 2013 [87]
Trichoderma harzianum1Meloidogyne incognitaISR and SARMartínez-Medina et al., 2017 [89]
1 plant growth-promoting fungi (PGPF).
Table 3. A list of PGPR commercialized as BCAs against PPN.
Table 3. A list of PGPR commercialized as BCAs against PPN.
Commercial ProductsPGPRApplicationsReferences
BioNemaGonTMBacillus firmusReduce nematode population and root infestation by nematodes in vegetables and herbs[90]
BioYieldTM Bacillus subtilis GB03, Bacillus amyloliquefaciensNematodes in tomato, strawberry, and bell pepper[65]
Clariva® pnPasteuria nishizawae Pn1Seed treatment; Target Heterodera glycines to reduce feeding and reproduction, and increase yields under heavy PPN pressure. [91]
Deny, Blue CircleBurlkholderia cepaciaInhibit egg hatching and mobility of nematode juveniles[92]
MeloCon®, BioAct and NemOutPurpureocillium lilacinus 251Inhibit root knot, burring, cyst, reniform, spiral, sting, and root lesion nematodes. [93]
Naviva ST Pasteuria sp. Ph3Seed treatment; Inhibit Rotylenchulus reniformis in cotton, soy, vegetables, cucurbits, and floriculture. [94]
NewProPasteuria usgae Bl1 + Pasteuria sp. Ph3 Inhibit lance and sting nematodes in turf (Bermudagrass and St. Augustine grass) [95]
Nortica 10 WPBacillus firmus I-1582Inhibit cyst, lance, lesion, ring, root knot sheath, spiral, sting, and stunt nematodes in turf.[96]
VOTiVO FS Bacillus firmus I-1582Seed treatment; inhibit a broad range of nematodes. Available also as premix with insecticide [97]
Table 4. A list of PPN inhibitory PGPR with uncharacterized function.
Table 4. A list of PPN inhibitory PGPR with uncharacterized function.
PGPRTarget PPNTarget CropsReferences
Alcaligenes faecalisMeloidogyne incognitaChickpeaSiddiqui and Mahmood, 1992 [22]
Azotobacter chroococcumMeloidogyne incognita, Meloidogyne javanicaEggplant, TomatoBansal et al., 2002 [98]
Bacillus altitudinis,
Bacillus aerophilus,
Bacillus aryabhattai,
Bacillus galliciensis,
Bacillus psychrosaccharolyticus,
Bacillus safensis,
Bacillus siamensis,
Bacillus simplex,
Bacillus toyonensis,
Bacillus weihenstephanensis
Meloidogyne incognitaCottonXiang et al., 2016 [66]
Bacillus firmusBelonolaimus longicaudatusBermudagrassCrow, 2014 [99]
Bacillus flexusMeloidogyne incognitaBasilTiwari et al., 2017 [100]
Bacillus isolatesHeterodera cajani, Meloidogyne incognitaPigeon peaSiddiqui and Shakeel, 2007 [101]
Bacillus methylotrophicusMeloidogyne incognitaTomatoZhou et al., 2016 [102]
Bacillus polymyxaMeloidogyne incognitaTomatoKhan and Akram, 2000 [103]
Bacillus tequilensisMeloidogyne incognitaBasilTiwari et al., 2017 [100]
Burkholderia cepaciaMeloidogyne incognitaTomatoMeyer et al., 2000 [104]
Lysinibacillus sphaericusMeloidogyne incognitaTomatoColagiero et al., 2018 [105]
Lysobacter spp.Meloidogyne incognitaTomatoZhou et al., 2016 [102]
Paenibacillus lentimorbus, Paenibacillus polymyxaMeloidogyne incognitaTomatoSon et al., 2009 [106]
Paenibacillus maceransMeloidogyne exiguaCoffeeOliveira et al., 2007 [107]
Pseudomonas solanacearumRotylenchulus reniformisEggplantKermarrec et al.,1994 [108]
Pseudomonas striataMeloidogyne incognitaPeaSiddiqui and Singh, 2005 [109]
Pseudomonas stutzeriMeloidogyne incognitaChickpeaSeenivasan et al., 2001 [110]
Stenotrophomonas maltophiliaParatrichodorus pachydermus, Trichodorus primitivusPotatoInsunza et al., 2002 [111]
Streptomyces spp.Meloidogyne incognitaEggplant, TomatoRashad et al., 2015 [112]

Share and Cite

MDPI and ACS Style

Subedi, P.; Gattoni, K.; Liu, W.; Lawrence, K.S.; Park, S.-W. Current Utility of Plant Growth-Promoting Rhizobacteria as Biological Control Agents towards Plant-Parasitic Nematodes. Plants 2020, 9, 1167. https://doi.org/10.3390/plants9091167

AMA Style

Subedi P, Gattoni K, Liu W, Lawrence KS, Park S-W. Current Utility of Plant Growth-Promoting Rhizobacteria as Biological Control Agents towards Plant-Parasitic Nematodes. Plants. 2020; 9(9):1167. https://doi.org/10.3390/plants9091167

Chicago/Turabian Style

Subedi, Pratima, Kaitlin Gattoni, Wenshan Liu, Kathy S. Lawrence, and Sang-Wook Park. 2020. "Current Utility of Plant Growth-Promoting Rhizobacteria as Biological Control Agents towards Plant-Parasitic Nematodes" Plants 9, no. 9: 1167. https://doi.org/10.3390/plants9091167

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