Skip to main content

Plants and endophytes interaction: a “secret wedlock” for sustainable biosynthesis of pharmaceutically important secondary metabolites

A Correction to this article was published on 02 January 2024

This article has been updated

Abstract

Many plants possess immense pharmacological properties because of the presence of various therapeutic bioactive secondary metabolites that are of great importance in many pharmaceutical industries. Therefore, to strike a balance between meeting industry demands and conserving natural habitats, medicinal plants are being cultivated on a large scale. However, to enhance the yield and simultaneously manage the various pest infestations, agrochemicals are being routinely used that have a detrimental impact on the whole ecosystem, ranging from biodiversity loss to water pollution, soil degradation, nutrient imbalance and enormous health hazards to both consumers and agricultural workers. To address the challenges, biological eco-friendly alternatives are being looked upon with high hopes where endophytes pitch in as key players due to their tight association with the host plants. The intricate interplay between plants and endophytic microorganisms has emerged as a captivating subject of scientific investigation, with profound implications for the sustainable biosynthesis of pharmaceutically important secondary metabolites. This review delves into the hidden world of the "secret wedlock" between plants and endophytes, elucidating their multifaceted interactions that underpin the synthesis of bioactive compounds with medicinal significance in their plant hosts. Here, we briefly review endophytic diversity association with medicinal plants and highlight the potential role of core endomicrobiome. We also propose that successful implementation of in situ microbiome manipulation through high-end techniques can pave the way towards a more sustainable and pharmaceutically enriched future.

Introduction

Almost all the living organisms on Earth interact with one another in different ways, and they all coexist as a community. Similarly, plants are interconnected and impacted by the presence of microbes both above and below ground, which ultimately play a critical role in the fitness of the host plants [1]. The term "plant microbiome" broadly refers to all distinctive microbial communities that inhabit the endosphere (plant internal tissues), phyllosphere (air-plant interface), and rhizosphere (plant roots-soil interface) [2, 3]. The endophytes form tight associations with their host and thus play a pivotal role in plant growth, fitness, and development in addition to protecting against biotic and abiotic stresses via secretion of indole-3-acetic acid (IAA), siderophores, phosphate solubilizers, etc. [4,5,6]. In addition, some bacterial endophytes provide nitrogen to the host via nitrogen fixation [7].

In most cases, the interaction is mutually beneficial as the plant provides carbon in return for other nutrients, metabolites and protection against pests, pathogens and abiotic stresses, thereby altering the plant metabolome in various ways [8]. The bioactive secondary metabolites are derived from intermediate compounds of primary metabolism, and they are not directly required for the organism’s basic growth and development [9]. Although the precise roles of secondary metabolites in plant metabolism and physiology are not fully known, it is thought that they participate in a variety of interactions between plants and their environment, such as protection from biotic and abiotic stressors, pollinator attractants, signaling molecules etc. and thus offer a selective advantage to the sessile plants [10,11,12,13]. Additionally, once viewed as waste products, these secondary metabolites hold immense pharmaceutical importance for human health [14].

The enormous diversity of secondary metabolites in medicinal plants is gaining more recognition as a valuable reservoir of novel chemical compounds that exhibit diverse pharmacological effects. These metabolites have been isolated from higher plants in amounts close to 100,000, where medicinal plants hold a major share [15]. In the past few decades, with the advancement in science, the concept that plants individually produce metabolites has changed, and the role of microbes in modulating the metabolites has been increasingly documented [16,17,18,19]. In this regard, the ability of the endophytes to produce active compounds appears to be integral to their functions, as recent studies have demonstrated that endophytes influence the production of host secondary metabolites through a number of biochemical processes [15].

Most of the studies involving endophyte plant interactions have majorly focused on yield-enhancing attributes with less focus on how these microbes alter the plant chemistry, particularly the biosynthesis of secondary metabolites [20,21,22]. Nevertheless, few studies have been conducted in various non-medicinal plants, including tomato [23], rice [24, 25], soybean [26] and grapevine [27], where the impact of endophytes on secondary metabolite biosynthetic pathways have been elucidated with regard to biotic and abiotic stress tolerance mechanism. However, given that medicinal plants are highly recognised for their pharmacological attributes, a demand fueled by the pharmaceutical, herbal medicine, and nutraceutical industries, it becomes imperative to delve deeper to understand their role as it might lead to drug discovery, promote sustainable agriculture and can have wider applications in the field of medicine, environmental science, and biotechnology [28].

Thus, the focus of this article is primarily to comprehend the mechanism and role of endophytes in fine-tuning the reservoir of incredibly diverse pharmaceutically functional compounds, especially in medicinal plants. We believe that a more comprehensive understanding in this regard can be effectively utilized and harnessed for developing superior and more potent drugs derived from medicinal plants. Additionally, through this review, we have also highlighted the potential role of “core endophytes” in secondary metabolites enhancement that could be game changers in time to come. Finally, we propose that integrating multiomics approaches may help design a “customized bioformulation” of metabolites in the near future.

Medicinal plants: gold mine of bioactive secondary metabolites

Plant metabolites are low molecular weight organic compounds classified into three main categories. This includes the primary metabolites that are usually highly conserved and directly required for the growth of plants. The second category consists of secondary metabolites comprising major groups of phenolics, terpenes, and nitrogen-containing compounds, and the third is the plant hormones that regulate organismal processes and metabolism of plants [29]. The documented plant metabolome comprises approximately over 2,000,000 characterized secondary metabolites in the plant kingdom with multidimensional applications, which majorly belong to categories like tannins, alkaloids, phenols, glycosides, volatile oils, saponins, resins, steroids, terpenoids, and bitter principles [30,31,32]. Among the diverse group of plants, medicinal plants are the “goldmines” of an extensive assortment of phytopharmaceutically important bioactive molecules. This is why these plants have been utilized for restoring and maintaining health, preserving and flavoring food, enhancing daily life with color and aroma, and opening doors to mystical experiences and spiritual dreams since the dawn of humanity [33]. Medicinal plants biosynthesize and accumulate a diversified class of secondary metabolites in sufficient extractable form to be economically utilized as primary resources for various commercial, scientific or technological applications [34]. Usually, the biosynthesis of primary and secondary metabolites occurs along the same pathway. The excess carbon produced during primary metabolism is essentially absorbed and stored in the form of secondary metabolites, which in time of need, are disintegrated and used again in primary metabolism, thereby maintaining the perfect balance between primary and secondary metabolic pathways in the plants [10]. Although the secondary metabolites do not take part in the plants' primary growth and development, they serve various roles in plants, from serving as defense mechanisms against pathogens, attracting pollinators and as chemical signals to other plants [33].

Bioactive molecules produced by the plant possess broad range of pharmacological and therapeutic potentialities such as antimicrobial, anticancer, antioxidant, antiviral, antitumor, anti-inflammatory, hepatoprotective, antidepressant, antidiabetic, antiatherosclerotic, antithrombotic, vasoprotective, memory enhancer, cardiovascular improver, anti-AIDS, anti-Parkinson’s disease, anti-Alzheimer’s, anti-cognitive impairment and immunoprotective effects [33, 35]. In fact, 40% of human medicine originates from natural sources, of which 25% and 13% are majorly contributed by plants and microbes, respectively [36]. For example, many plant-based novel drugs such as paclitaxel, toptecan, teniposide, ectoposide, plaunotol, vinblastin, z-guggulsterone, gomishin, nabilone, lectinan, artemisinin are being used globally for the wellbeing of the humans. The remarkable contribution of medicinal plants can be well validated by citing examples like the serpentine compound obtained from the roots of Rauwolfia serpentina, which is known for its anti-hypertension effect on the body. Similarly, vinblastine from Catharanthus roseus is applied to treat neck cancer, Hodgkins and choriocarcinoma [37]. The medicinal plants are also home to a variety of bioactive components which have an antineoplastic effect, such as vermodalin (Vernonia amygdalina), chebulinic acid, ellagic acid, and tannic acid (Terminalia chebula) and allicin (Allium sativum) [38].

The significant pharmaceutical contribution of several medicinal plants belonging to families like Liliaceae, Asteraceae, Rutaceae, Apocynaceae, Solanaceae, Piperaceae, Caesalpinaceae, Sapotaceae, Ranunculaceae, Apiaceae are considered potential sources of many bioactive compounds with direct application in pharmaceutical industries. It is believed that the immense biosynthetic potential of medicinal plants has not yet been fully unveiled, and it is proposed that leveraging the most recent advancements in technologies like microbiome engineering and gene editing may produce unique chemical compounds with improved or novel bioactivities.

Diversity and distribution of endophytes in the medicinal plants

Microbial endophytes are known to be associated with all plant species, ranging from perennial trees and medicinal plants to various other crops [39, 40]. Endophytes inhabit various plant parts, including roots, leaves, stems, flowers, and seeds. The composition, richness, and population of endophytic microbial communities differ based on factors such as plant species, the specific plant compartments (e.g. roots, stems and leaves), plant age, and surrounding environmental conditions [1, 41, 42]. Evidence suggests that endophytes are acquired vertically (via seed) and horizontally from the plants soils where plant grow. Most of these endophytes are acquired from the soil by active filtration and mutual recognition between plant hosts and microbes. Plants secrete chemoattractants in various forms through root exudates to attract specific microbes. Only those microbes that can recognize those plant chemoattractants can migrate to the plant tissues, recognise the host, and penetrate the plant surface. The majority of endophytes colonize the intercellular spaces; however, many times, they are also found to be colonizing the intracellular spaces and vascular system of the plants [43].

Additionally, only endophytes with specific traits like the presence of plant polymers breakdown machinery, protein secretion system, redox response regulatory systems, quorum sensing, etc., successfully colonize and inhabit the plant endosphere, which is a relatively small space [44]. Furthermore, the interaction between the plant immune system and microbial recognition significantly impacts the successful colonization of plant tissues by microbes [1]. Continuous and progressive filtering means endophyte diversity declines from root to leaf to flower and seeds. Some microbes are shared between different compartments, while others are specialised for a particular compartment/niche (e.g. leaves). A greater understanding of the microbial makeup and abundance is now attainable because of the advancements in sequencing technology, which have also made it possible to decode the whole endophytic variety of organisms living inside the plant tissues. Compared to the high throughput next-generation sequencing technologies, traditional culture-dependent methodologies offer far less information on the variety of microorganisms. However, the direct examination of microbial activity and its interactions with the host is an additional benefit of conventional techniques [45]. The information available through modern sequencing and traditional technologies reveals the presence of both prokaryotic and eukaryotic groups of microorganisms in the plant system [4]. Amongst the prokaryotic group, most of the endophytic bacteria are distributed among four bacterial phyla, namely Proteobacteria, Firmicutes, Actinobacteria and Bacteroidetes, with major dominant genus as Pseudomonas, Streptomyces, Bacillus, Serratia, Micrococcus, Burkholderia, Enterobacter, Mycobacterium, Rhizobium etc. [4, 19, 46].

Similarly, endophytic fungi too are found to be active colonizers of the plant tissues, with Ascomycota predominating over 95% of the endospheric population of the plants, followed by Basidiomycotas [47, 48]. The dominance of ascomycetous fungi such as Penicillium, Curvularia, Cladosporium, Aspergillus, Colletotrichum, Trichoderma, etc., has been documented in the plant endosphere by various research groups [48, 49]. The detailed list of endophytic diversity associated with the various plant parts of medicinal plants with their respective plant growth-promoting activity has been mentioned [19, 50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107] in Additional file 1: Table S1.

Rooted bonds: unravelling the early colonization events in plant-endophyte interaction

The exact mechanisms responsible for the relationship between plants and endophytes are still poorly understood. However, studies to date suggest that endophytic microbes, like any other microbe, share characteristic features such as rhizosphere competence, motility trait, protein-enzyme secretion systems and an ability to overcome plant immunity [108]. Due to selective pressure, the colonization of endophytic plant partners is organ and tissue-specific, ranging from roots to shoots, shoots to flowers, flowers to fruits and finally, many times to the seeds [109, 110]. According to Kandel et al. [43], successful colonization involves the deployment of endophytes by the host plant near its vicinity, followed by the attachment of the endophytes to the host plant's surface and, finally, their entry into the plants [43].

The primary step of colonization begins with the recruitment of endophytes towards the roots of the host plant with subsequent migration to stems and leaves through the xylem vessels, a process influenced by the rich exudates released by plants into the rhizosphere [111, 112]. Researchers have previously demonstrated that bacterial endophytes possess chemotactic abilities, enabling them to swim toward the plant's root system by detecting root-secreted molecules [109, 113]. A study carried out in support of bacterial chemotaxis revealed maximum utilization of the root exudates by endophytic Stenotrophomonas maltophila RCT31, which ultimately resulted in enhanced plant growth of a medicinal legume Clitoria ternatea L. [114]. Likewise, fumaric acid released by Panax notoginseng significantly stimulated the chemotaxis ability, growth and biocontrol-related genes of an endophytic Bacillus amyloliquefaciens subsp. plantarum YP1 [115].

Upon reaching the plant, bacterial endophytes employ various structural components like pili, flagella, and fimbriae, along with secretory products such as lipopolysaccharide (LPS) and exopolysaccharides (EPS), for adhering to the surface. These microbial appendages serve as propellers, translocating bacteria toward the plant's surface and overcoming repulsive obstacles [116]. In contrast, the recognition and attachment of specific hosts by fungal endophytes involve the release of fluids facilitating germling assemblage necessary for penetration. Transcriptional studies have revealed that fungal endophytes like Piriformospora indica secrete small secreted protein (SSP) effectors and disrupt phytohormone homeostasis during early symbiosis [117]. Upon successful attachment to the plant surface, endophytes find entry points through various plant structures, such as elongation zones, root hairs, cotyledons, stems, leaves, and flowers [118]. Passive penetration occurs through cracks, root tips, lenticels, stomata, and hydathode openings [119], while active entry is facilitated by molecules like EPS, LPS, quorum sensing signals, and chitin [21]. To avoid triggering plant resistance, endophytic microbes produce fewer cell wall-degrading enzymes and maintain lower cell densities to avoid getting detected as pathogens [120, 121]. Endophytes further utilize the xylem vessels for upward translocation, capitalizing on the low nutrient concentration in these vessels. Studies on Paraburkholderia phytofirmans PsJN revealed entry through the root exodermis into cortical cells via the endodermal layer [122], while the active role of alcohol dehydrogenases was observed in Azoarcus sp. BH72 colonization in waterlogged rice [123]. Likewise, dark septate endophytes (DSE) have been observed in fungal interactions that form alliances with plants through the root cortex, similar to arbuscular mycorrhizal fungi, employing strategies to maintain symbiosis [124]. Similarly, a unique strategy was observed in Piriformospora indica that secreted extracellular hydrolyzing enzyme adenosine 5'-triphosphate (eATP) in the host plant’s apoplast during later stages of interaction [125]. These findings illustrate the diverse and intricate mechanisms endophytes employ to colonize plants, shedding light on the complexity of these interactions. A diagrammatic illustration representing the early colonization events happening at plant endophyte interaction interphase has been represented in Fig. 1

Fig. 1
figure 1

Schematic illustration depicting routes adopted by endophytes for successful colonization in the host plant. A Recruitment mechanism of potential endophytes and its resultant diversity gradient in the host plant. B Crosstalk between host plant and endophytes leads to host plant nutrient and defence management. root exudates (REs), N-Acyl homoserine lactone (AHL), volatile organic compounds (VOCs), siderophore (Sid), secondary metabolites (SMs), indole-3-acetic acid (IAA), cytokinin (CK), gibberellic acids (GAs), reactive oxygen species (ROS), 1-aminocyclopropane-1-carboxylic acid (ACC), jasmonic acid (JA), ethylene (ET), salicylic acid (SA), pathogenesis-related (PR), type III secretion system (T3SS), type IV secretion system (T6SS), microbe-associated molecular patterns (MAMPs), quorum sensing (QS)

In planta secondary metabolite enhancement by the endophytes in medicinal plants

Recent studies reveal the enormous impact of the plant microbiome, including epiphytic and endophytic microbiomes, on the plant's overall health and performance [126]. Plant microbiomes are dynamic and can rapidly adapt to biotic, abiotic, and other environmental pressures [127, 128]. This can help host plants produce bioactive compounds at a steady rate. In addition, endophytes can mediate the re-configuration of the plant metabolome that depends on the combination of plant species/cultivar and other interacting microbial partners. Increasing evidence suggests that the interactions between the plant and the endophytic microorganisms increase the production of secondary metabolites such as alkaloids, flavonoids, and terpenoids in medicinal plants [129,130,131,132,133] (Fig. 2). These interactions in most cases are mutually beneficial, and a number of medicinal plants produce bioactive secondary metabolites that have significant direct and indirect impacts on the population and physiological processes of endophytic microbiota [134]. In addition, numerous research studies have indicated that root exudates play a significant role in shaping the rhizomicrobiome, affecting root-microbe interactions and endophytic diversity [135, 136]. In one study, the fungal communities associated with grapevines were influenced by the age of the leaves, with younger leaves exhibiting higher endophytic fungal diversity and richness compared to mature leaves [137]. However, the processes by which endophytes encourage the production of secondary metabolites in plants have been the subject of several investigations without a common consensus. In one such investigation, endophytes of Panax ginseng were found to convert ginsenosides into different forms that ultimately influenced the efficacy of the host plant [138]. It is postulated that endophytes' secondary metabolite enhancement in planta is linked with plants' encouragement to accumulate more photosynthetic material. This promotes the upregulation of the genes involved in the plant secondary metabolite biosynthesis pathway, changes in the genetic makeup of the plants or distinctive metabolites produced by the endophytes impacting the plant biosynthetic pathways [139].

Fig. 2
figure 2

Endophyte mediated enhanced production of secondary metabolites and its applications. A Depicts plants inoculated with beneficial endophytes, significantly improving plant growth and development over non-inoculated plants. The zoomed-out leaf portion represents the modulation of host plant secondary metabolic pathways by multiple transcription factors regulated by upstream signals in response to endophytic colonization. B Different categories of bioactive secondary metabolites (structures given alongside) and their multifaceted industrial applications. microbe-associated molecular patterns (MAMPs), transcription factors (TFs), secondary metabolite (SM), mevalonic acid (MVA), methylerythritol phosphate (MEP), acetoacetyl CoA thiolase (AACT), 3-hydroxy-3-methylglutaryl-CoA synthase (HMGS), 3-hydroxy-3-methylglutaryl-CoA reductase (HMGR), mevalonate kinase (MVK), phosphor mevalonate kinase (PMK), -hydroxy-3-methylglutary-CoA (HMG-CoA), 3 mevalonate5-diphosphate decarboxylase (MVD), isopentenyl diphosphate (IPP), dimethylallyl diphosphate (DMAPP), IPP isomerase (IDI), mevalonate pyrophosphate (MVPP), farnesyl-diphosphate synthase (FPS), famesyldiphosphate (FPP), geranyl phosphate synthase (GPS), geranyldiphosphate (GPP), geranyl diphosphate synthase (GPPS), geranyl geranyl diphosphate (GGPP), geranyl geranyl diphosphate synthase (GGPPS), 1-deoxy-D-xylulose5-phosphate synthase (DXS), 1-deoxy-D-xylulose5-phosphate (DXP), 1-deoxy-D-xylulose5-phosphate reductoisomerase (DXR), MEP cytidylyltransferase (MCT), 1-hydroxy-2-methyl-2-(E)-butenyl4-diphosphate synthase (HDS), MEP-cPP synthase (MCS), squalene synthase (SQS), cytochrome P450 reductase (CPR), sterol methyltransferase (SMT), squalene epoxidase (SQE)

Microbes also produce many metabolites for various purposes, including interacting with the host plant [140]. Since many metabolites are produced by microbes or by their interaction with the host plant, the focus is currently being shifted from the medicinal plants' bioactive secondary metabolites individually to linking the secondary metabolome of plants with the plant’s endomicrobiome [141]. This is because endophytic microbes are important in forming bioactive secondary metabolites such as steroids, alkaloids, polyketones, peptides, flavonoids, terpenoids, and phenols [142, 143]. Similarly, some fungi, like Curvularia sp. and Choanephora infundibulifera, are known to increase the level of vindoline, a terpenoid indole alkaloid (TIA), in the leaves of endophyte-free Catharanthus roseus plants by 403% and 229%, respectively. The research conducted by Pandey et al. [144] yielded molecular evidence indicating an increase in the expression of genes responsible for both the structural and regulatory aspects of the TIA biosynthesis pathways in plants inoculated with endophytes [144]. Likewise, tanshionone biosynthesis was significantly increased in hairy root cultures upon application of the polysaccharide portion of an endophytic Trichoderma atroviride [145]. Similarly, an integrated study involving comparative metabolomics and transcriptomics of Epichloe festucae-infected and uninfected ryegrass plants reported the host metabolism reprogramming in favor of secondary metabolism over primary metabolism [146].

Many studies have reported endophytes-mediated modulation of plant metabolites in recent years. For example, an endophyte-mediated accumulation of forskolin content in Coleus forskholli was observed by the application of three indigenous endophytes of C. forskohlii namely Fusarium redolens (RF1), Phialemoniopsis cornearis (SF1) and, Macrophomina pseudophaseolina (SF2) [147]. The findings were validated by the expression study of five key forskolin biosynthetic pathway genes, namely CfTPS2, CfTPS3, CfTPS4, CfCYP76AH15, and CfACT1-8 [148]. Similarly, bacterial and fungal endophytes linked with the Agarwood tree (Aquilaria malaccensis) increased the production of agarospirol, a highly sought-after product in the pharmaceutical and fragrance industries [149]. Interestingly, tissue-specific localization of the endophytes was found to be linked with Papaver somniferum overall plant performance, as most leaf endophytes influenced plant development and productivity, while capsule endophytes played a key role in influencing alkaloid production [72]. Many other endophytic bacteria, such as Pseudomonas fluorescens, Azospirillum brasilense, Bacillus subtilis, Paenibacillus polymyxa, Stenotrophomonas and others, have also been reported to increase the production and accumulation of significant secondary metabolites in the host [11, 12, 16,17,18,19, 39, 129, 147, 149, 150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193] (Additional file 1: Table S2). We argue that we have only scratched the surface, and many secondary metabolites produced by plant-microbial interaction remain to be discovered and characterized. Theoretically, endophytes can impact plant metabolites' types, quantity and quality. We hypothesize that the endophytes can encourage plants for more primary and secondary metabolites through the secretion of microbial metabolites or other signal molecules, phytohormones, upregulation of defence pathways, epigenetics, etc. Additionally, they can also influence the quality of these metabolites by microbial degradation or conjugation between plant and microbial metabolites. At this point, the role of endophytic enzymes cannot be ignored, which might catalyze the initial steps of secondary metabolite biosynthetic pathways in the host plant by initiating, activating or inhibiting certain biosynthetic routes. There is also a high possibility that the endophytic microorganisms can harbor novel enzymes not present in the host plant, which may introduce new chemical reactions or biosynthetic pathways, leading to the synthesis of previously unknown secondary metabolites in the plant.

Can core endomicrobiome be a key player in the modulation of secondary metabolites biosynthesis in medicinal plants?

Broadly, the set of operational taxonomic units consistently connected and shared by microbial communities from various but related hosts that provide specific host functions is often referred to as the “core microbiome” (CM) [194, 195]. Core microbiomes of various plants have been identified and reported [196,197,198]. Recently, the concept of identifying functionally important microorganisms that consistently associate with a host species is being added to the prevailing concept of CM, which is majorly based on the idea that a taxon's persistence across the spatial and temporal boundaries of a particular habitat is directly reflective of its functional importance within the niche it occupies [199, 200]. The endosphere of the plants is reported to be colonized by the community of microbial taxa with desired functional attributes, which collectively form the “core endomicrobiome” (CEM). Some genera, like Bacillus and Pseudomonas, are ubiquitous and can be found across the entire plant system [46, 201]. Previous studies have highlighted the seeds' vertical transmission of such essential microbiota over generations [202, 203]. CEMs, because of their constancy and uniformity, are thought to be essential for host fitness and hence have the ability to modulate plant microbiomes to achieve desired outcomes [204, 205]. These CEMs could be mutualists, commensalist, or occasional antagonists who, apart from directly influencing the secondary metabolite biosynthesis, might also act as key modulators for the secondary metabolite level [206]. The composition and role of CEMs have been obtained for various plants such as Arabidopsis, tomato, and citrus. However, limited research has been conducted to establish connections between the potential beneficial traits of the CEMs and their functions. For example, the CEMs analysis of Oryza sativa leaves divulged that Acinetobacter, Enterobacter, Pseudomonas, Pantoea, Sphingomonas, Stenotrophomonas, and Rhizobium genera were found to be a part of the core microbiome [207]. Likewise, Bulgarelli et al. [208] and Lundberg et al. [209] defined the microbial community structure and core endophytic population of the Arabidopsis root microbiome, which showed the dominance of Actinobacteria followed by Proteobacteria, Bacteroidetes and Firmicutes [208, 209]. Similar outcomes were also observed in the core-endophytic community analysis of tomato and sugarcane roots [210,211,212,213].

Since it is evident that medicinal plant endophytic microbial assemblage influences the synthesis of secondary bioactive metabolites directly or indirectly by regulating the plant metabolic pathways [214,215,216], the role of CEMs in this context needs further exploration. In one of the earliest studies carried out on deciphering the CEM of medicinal plants, Wicaksono et al. [217] provided valuable insights into the fact that distinct plant organs of Leptospermum scoparium are capable of co-harboring specific core microbial taxa that affect the bioactive properties of the plant [217]. Likewise, some signature key core microbes were also identified from two medicinal plants, namely Achillea millefolium L. and Hamamelis virginiana that were hypothesized to be the key modulators of the metabolome of the plants [48].

In another investigation, Echinacea purpurea has been proposed to be an excellent model herb for investigating microbiome-secondary metabolites interactions as endophytes of this plant modified the production of volatile compounds such as phenylpropanoid and alkamides [218]. Similarly, significant induction of enantiomeric naphthoquinones such as alkannin and shikonin by endomicrobiome in Alkanna tinctoria [16] and 19-tetrahydrocannabinol and cannabidiol in Cannabis sativa and morphine production in Papaver somniferum, were observed [215, 219]. Recently, in one of the studies, seed-associated CEMs were proposed to influence tanshinone production [220]. This postulation is based on the evidence that the identified CEMs comprise a gene reservoir associated with the terpenoid backbone synthesis, offering extra metabolic capabilities to Salvia miltiorrhiza. Working on the above line, our group has identified a set of core endomicrobiome genera, namely Pseudomonas, Paenibacillus, and Bacillus from Andrographis paniculata that, along with the introduced endophytic strain were speculated to contribute to the enhanced andrographolide content within the plant tissues [221].

Growing evidence suggests that these CEMs, in combination, probably enhance the plant’s physicochemical homeostasis by triggering secondary metabolite biosynthesis. However, there are still several gaps and challenges in this study area, as identifying effective endophytes for specific metabolite enhancement is difficult due to their vast diversity. Additionally, the ability of endophytes to enhance secondary metabolites can be species-specific and variable. Therefore, understanding the factors contributing to this specificity and variability is essential for reliable application. Complex host-endophyte interactions and the impact of environmental conditions also require further investigation for reliable applications. Despite observing shifts in endophyte diversity in response to plant metabolites, the functional consequences of these changes remain inadequately understood. Therefore, advanced genomic tools and a comprehensive understanding of endophyte biology and plant secondary metabolism are required to address these gaps and challenges.

Bridging the gap through microbiome engineering

Irrespective of the important roles played by the microbiome, the vast bulk of the transformative potential of the microbial world is yet to be discovered and applied. It is postulated that targeted manipulation of plant microbiome via inoculation with beneficial microbes and pre- and postbiotics that promote the production of secondary metabolites can potentially help produce industrial and economically viable quantities of target metabolites. However, such approaches have their own set of limitations and have been discussed in detail in agricultural contexts [139, 222]. Recently, approaches to overcome such limitations were proposed, including explicit consideration of theoretical framework in inoculant testing and in situ microbiome manipulations/engineering [42, 222]. In this regard, modulating the plant holobiont through microbiome engineering is an emerging biotechnological approach for increasing crop yields, resilience and secondary metabolite biosynthesis in plants [223, 224].

The preliminary aspect of microbiome engineering is identifying the core microbiome and host functions provided by individual members and their interaction. This is followed by targeted manipulation of plant microbiome composition to achieve specific phenotypes, in this case, high quantity and quality of secondary metabolites for pharmaceutical and other applications. However, our current knowledge and ability to manipulate microbiomes are limited. We need systematic studies first to identify the interactive impact of plant microbiomes and then identify signal molecules (including volatiles) that trigger metabolite synthesis and microbial modification. This will require the utilization of manipulative work in combination with combined multi-omics techniques like metagenomics, metaproteomics, and metatranscriptomics to determine the functionality of the entire microbial community within a particular niche [225, 226] (Fig. 3).

Fig. 3
figure 3

Top-down and bottom-up approaches to engineer microbiomes. A The panel represents the bottom-up design methodology that begins with isolates. Genome editing boosts system functions by recognizing gene editing sites that reroute metabolic flux to the intended secondary metabolites. B The panel represents the top-down design methodology. a Depiction of native endophytic micriobiome hub with interacting partners. Categories of the hub endomicrobiome are exclusively based on network topology information within a microbial network. The possible roles of secondary metabolites (SM) inducing core endomicrobiome (CEM) that are expected to mediate interactions between plants and native microbial species includes (i) Scoring of CEMs based on enrichment of SM inducing microbial species in the rhizosphere through chemoattractants released in the form of root exudates. (ii) Restrainment of deleterious biotic stressors. (iii) Bolstering of SM inducing functional CEMs by applying facilitative microbiomes. b Preparation of endosymbiotic cells derived from plant samples. (i) Surface sterilization of plant tissue for removal of epiphytic microbes. (ii) Sample crushing followed by gradient centrifugation for concentrating endomicrobiome. (iii) Isolation of single cell microbial droplets with CEMs using microdroplet devices (iv) Compartmentalization of single cell CEMs using microfluidic devices to separate cells from microbial mixtures. (v) Cell sorting using vibrational spectroscopy techniques like surface enhanced Raman scattering (SERS)/tip enhanced Raman scattering (TERS) and Fourier transform infrared (FTIR) (vi) Optimization of culture media for isolating CEM/s with varying nutritional requirements and growth conditions. (vii) Library development of all possible CEMs using permutations and combinations (viii) High throughput inoculation of sets of CEMs in seeds/seedlings/plants (ix) Identification of potential SM inducing CEMs by chemical profiling of plant tissue/s (x) Mass multiplication of potential SM inducing CEMs for large scale cultivation with the ultimate aim of increasing microbial heterogeneity. The middle panel represents the key aspects of microbiome engineering with increasing and decreasing complexities in bottom-up and top-down approaches. The various shapes of the microbes indicate different isolates chosen throughout the designing process

Identification of signal molecules using sensitive instruments (e.g. GC–MS, HPLC–MS) that trigger plant synthesis and/ or microbial modification can be used to manipulate in situ microbiomes in future. Recently, a four-step cycle was proposed to enable efficient microbiome engineering: design-build-test-learn (DBTL) [227]. This cycle entails creating an initial microbiome design to accomplish a specified engineering goal, building the microbiome, testing its functionality against a set of predetermined metrics to see if the design-build solution(s) produced the designed objective, understanding the outcomes and shortcomings, and utilizing new information for the upcoming DBTL cycles. By following this cycle iteratively, researchers can make step-by-step improvements, enhance the endophyte-host interaction, and achieve targeted enhancement of specific secondary metabolites in the host plant.

The top-down and bottom-up approach to design microbiomes

Empirical-based attempts to pinpoint the specific functions of individual components in producing community-derived phenotypes are difficult to accomplish due to biology's nonlinearity and the functional complexity present in microbial communities [228]. Therefore, to address the intricacy and to create biotechnological applications, top-down and bottom-up approaches can be adopted to design “customized endomicrobiome”. The top-down design enables the user to select various environmental parameters, which in turn enforces the existing microbiome to remodel itself and modulate to exhibit the desired biological functions [229]. For implementing a top-down approach, microbial resource management is an effective way of predicting the process of manipulating an ecosystem, which requires researchers to intellectualize a system that takes into account different inputs and outputs considering physiological parameters such as pH, redox potential, temperature, humidity etc. and predicts their capabilities to promote or restrict the particular biological process of interest [230]. Whereas, in the bottom-up approach, systems are pieced together to create increasingly sophisticated systems, making the initial systems subsystems of the budding system. When we move from DNA components to microbial ecosystems, this approach requires understanding how the interacting metabolic networks of each microorganism to be incorporated into the microbiome may affect the desired output [223, 227]. This will require a design process involving genomes of key microbiome members [231], reconstructing their metabolic networks [232], and the use of modelling tools [233] and/or network analysis tools [234] to finally come up with a best-fit design. Overall, the bottom-up engineering approach is supported by logical guidance and requires experienced practitioners with the requisite foundation for wise engineering decision-making since all implementation depends on prior knowledge. This approach can be well adapted for systematically enhancing secondary metabolites through endophytic plant partners in controlled conditions. We believe that if the study is precisely designed and constructed to engineer a microbiome, achieving targeted and reliable enhancement of specific secondary metabolites in the host plant is not a very far-off dream.

Assembling microbiomes for achieving the desired traits in the host plant

New inventive approaches are needed to recruit beneficial microbes that positively impact plant performance. A theoretical network framework was proposed to identify optimal CMs recruited in a microbial network at central positions with associated extra microbes encompassing desired functions for the benefit of the hosts [206]. We thrust upon that utilizing the same approach can potentially enhance secondary plant metabolites production. For example, CEM in medicinal plants can be manipulated/ engineered in two ways: by self-assembly or by synthetic approach. Self-assembled microbiomes can be developed as open consortia using bio-reactors or biostimulators in which the building process actualizes an atmosphere conducive to the growth and desirable activity of the selected microorganisms [227]. Though building up microbiomes by applying axenic microbial cultures results in less complexity, designing synthetic microbiomes is difficult because of significant knowledge gaps about the taxa that are unculturable, uncharacterized and genetically unavailable with potentially substantial roles in agriculture applications. Therefore, to bridge the gap, innovative separation and controlled microbiome assembly methods like single-cell sorting [235] in combination with progressive culturomics [236] and phenotyping [237] can be used to encapture and employ the uncharacterized class of the metabolically diversified community.

Further, a designer assembly can be potentially attained if the latest cell phenotyping and sorting techniques are considered. Raman-activated cell sorting (RACS) is an emerging technology that allows sorting and phenotypic characterization of cells based on their intrinsic biochemical profiles without needing external labelling [238]. Another technique, namely bio-orthogonal non-canonical amino acid tagging (BONCAT), offers an additional approach to analyze microbial anabolic activity in situ, which could be coupled with cell sorting to separate active cells from complex samples and further identify them by DNA sequencing [239]. After the cells are individually sorted, culturing through current techniques requires sophisticated setups occupying large spaces. However, advancement in culturomics has introduced microfluidic devices such as microfluidic chips, also known as lab-on-a-chip technology [240,241,242], which can be further used to create and modify micro-droplets that can facilitate close analysis of axenic cultures with relatively low reagent consumption, elimination of undesired microbial species followed by sequencing, characterization and phenotyping through multi-omics approaches [243]. Moreover, they also offer an additional ability to control microbial communities' density, shape and size [241] with an analysis rate of 100,000 microbiomes/day. Overall, even though no single study has been carried out in the case of medicinal plants in this regard, we firmly believe that information from databanks and associated informatics tools offering the functionality of microbes to the annotated network data can be of great help in designing CEMs for the introduction into medicinal plant endosphere for successfully modulating the plant’s chemistry.

Conclusion, prospects and key questions

Endophytes play important roles in manipulating the secondary metabolite biosynthetic pathways with pharmaceutical and other chemical applications. Increasing quality and quantity of secondary metabolite productions are high and demand, and we propose that leveraging the core endomicrobiome of medicinal plants can provide effective solutions. However, owing to the complexity and multitude of interactions between microbiome-plant and the large diversity of plant microbes that remains poorly described, the precise engineering of CEMs for producing secondary metabolites will need concerted planning and collective efforts. The emerging tools of omics combined with the application of microfluidics, synthetic biology, genome editing, machine learning and data designing provide an opportunity to carry out large-scale endomicrobiome and plant/microbial metabolite screening. The success of such approaches will require transdisciplinary approaches involving active collaborations among agriculturists, chemists, engineers, microbiologists, plant molecular biologists/biochemists, and big data analysts. Based on the recent advancements in research tools and techniques, there is an opportunity to transform the production of secondary metabolites in medicinal plants by harnessing core microbiomes and plant-microbiome interactions through sustained resourcing and systems-based approaches (Fig. 4). In addition, we believe that by addressing some important key questions given below, the lacunas in the endophyte plant secondary metabolite enhancement can be bridged.

  1. 1.

    Are there any specific core endophytic species or strains consistently associated with all the medicinal plants in general that hold promise for discovering new drugs or therapeutic agents?

  2. 2.

    Considering their vast diversity, how can effective endophytes be identified for specific metabolite enhancement?

  3. 3.

    How do endophytes establish and maintain their interactions with medicinal plants, which are rich reservoirs of antimicrobial compounds?

  4. 4.

    Have the medicinal plants and their associated endophytes coevolved over time, and can investigating the drivers of host specificity and the ecological factors provide insights into the evolution of these interactions?

  5. 5.

    What factors contribute to species-specific and variable abilities of endophytes to enhance secondary metabolites?

  6. 6.

    How do host-endophyte interactions and environmental conditions influence the efficacy and reliability of endophyte applications?

  7. 7.

    Can understanding the functional variation among different endophyte taxa and their effects on host plants help harness their potential for medicinal plant cultivation?

  8. 8.

    How can we better understand the functional consequences of shifts in endophyte diversity in response to plant metabolites?

  9. 9.

    How can advanced genomic tools and approaches effectively address the gaps and challenges in studying endophytes and plant secondary metabolism?

Fig. 4
figure 4

Schematic representation of the complex dynamics of plant-endophyte interactions: Harnessing the power of endomicobiome engineering and mutiomics insights for its practical applications

Availability of data and materials

Not applicable.

Change history

Abbreviations

CM:

Core microbiome

CEM:

Core endomicrobiome

References

  1. Trivedi P, Leach JE, Tringe SG, Sa T, Singh BK. Plant–microbiome interactions: from community assembly to plant health. Nat Rev Microbiol. 2020;18(11):607–21. https://doi.org/10.1038/s41579-020-0412-1.

    Article  CAS  PubMed  Google Scholar 

  2. Schlaeppi K, Bulgarelli D. The plant microbiome at work. Mol Plant Microbe Interact. 2015;28(3):212–7. https://doi.org/10.1094/MPMI-10-14-0334-FI.

    Article  CAS  PubMed  Google Scholar 

  3. Compant S, Samad A, Faist H, Sessitsch A. A review on the plant microbiome: ecology, functions, and emerging trends in microbial application. J Adv Res. 2019;19:29–37. https://doi.org/10.1016/j.jare.2019.03.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hardoim PR, Van Overbeek LS, Berg G, Pirttilä AM, Compant S, Campisano A, Döring M, Sessitsch A. The hidden world within plants: ecological and evolutionary considerations for defining functioning of microbial endophytes. Microbiol Mol Biol Rev. 2015;79(3):293–320. https://doi.org/10.1128/MMBR.00050-14.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Le Cocq K, Gurr SJ, Hirsch PR, Mauchline TH. Exploitation of endophytes for sustainable agricultural intensification. Mol Plant Pathol. 2017;18(3):469–73. https://doi.org/10.1111/mpp.12483.

    Article  PubMed  Google Scholar 

  6. Dwibedi V, Rath SK, Joshi M, Kaur R, Kaur G, Singh D, Kaur S. Microbial endophytes: application towards sustainable agriculture and food security. Appl Microbiol Biotechnol. 2022;106(17):5359–84. https://doi.org/10.1007/s00253-022-12078-8.

    Article  CAS  PubMed  Google Scholar 

  7. Santi C, Bogusz D, Franche C. Biological nitrogen fixation in non-legume plants. Ann Bot. 2013;111(5):743–67. https://doi.org/10.1093/aob/mct048.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Etalo DW, Jeon JS, Raaijmakers JM. Modulation of plant chemistry by beneficial root microbiota. Nat Prod Rep. 2018;35(5):398–409. https://doi.org/10.1039/C7NP00057J.

    Article  CAS  PubMed  Google Scholar 

  9. Elshafie HS, Camele I, Mohamed AA. A comprehensive review on the biological, agricultural and pharmaceutical properties of secondary metabolites based-plant origin. Int J Mol Sci. 2023;24(4):3266. https://doi.org/10.3390/ijms24043266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Thirumurugan D, Cholarajan A, Raja SSS, Vijayakumar R. An introductory chapter: secondary metabolites. London: Intechopen; 2018. https://doi.org/10.5772/intechopen.79766.

    Book  Google Scholar 

  11. Xu W, Jin X, Yang M, Xue S, Luo L, Cao X, Zhang C, Qiao S, Zhang C, Li J, Wu J. Primary and secondary metabolites produced in Salvia miltiorrhiza hairy roots by an endophytic fungal elicitor from Mucor fragilis. Plant Physiol Biochem. 2021;160:404–12. https://doi.org/10.1016/j.plaphy.2021.01.023.

    Article  CAS  PubMed  Google Scholar 

  12. Ye HT, Luo SQ, Yang ZN, Wang YS, Ding Q, Wang KF, Yang SX, Wang Y. Endophytic fungi stimulate the concentration of medicinal secondary metabolites in houttuynia cordata thunb. Plant Signal Behav. 2021;16(9):1929731. https://doi.org/10.1080/15592324.2021.1929731.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jan R, Asaf S, Numan M, Kim KM. Plant secondary metabolite biosynthesis and transcriptional regulation in response to biotic and abiotic stress conditions. Agronomy. 2021;11(5):968. https://doi.org/10.3390/agronomy11050968.

    Article  CAS  Google Scholar 

  14. Jacobo-Velázquez DA, González-Agüero M, Cisneros-Zevallos L. Cross-talk between signaling pathways: the link between plant secondary metabolite production and wounding. Sci Rep. 2015;5:8608. https://doi.org/10.1038/srep08608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gandhi SG, Mahajan V, Bedi YS. Changing trends in biotechnology of secondary metabolism in medicinal and aromatic plants. Planta. 2015;241:303–17. https://doi.org/10.1007/s00425-014-2232-x.

    Article  CAS  PubMed  Google Scholar 

  16. Rat A, Naranjo HD, Krigas N, Grigoriadou K, Maloupa E, Alonso AV, Schneider C, Papageorgiou VP, Assimopoulou AN, Tsafantakis N, Fokialakis N, Willems A. Endophytic bacteria from the roots of the medicinal plant Alkanna tinctoria tausch (Boraginaceae): exploration of plant growth promoting properties and potential role in the production of plant secondary metabolites. Front Microbiol. 2021;12:633488. https://doi.org/10.3389/fmicb.2021.633488.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Singh S, Pandey SS, Shanker K, Kalra A. Endophytes enhance the production of root alkaloids ajmalicine and serpentine by modulating the terpenoid indole alkaloid pathway in Catharanthus roseus roots. J Appl Microbiol. 2020;128(4):1128–42. https://doi.org/10.1111/jam.14546.

    Article  CAS  PubMed  Google Scholar 

  18. Tripathi P, Tripathi A, Singh A, Yadav V, Shanker K, Khare P, Kalra A. Differential response of two endophytic bacterial strains inoculation on biochemical and physiological parameters of Bacopa monnieri L. under arsenic stress conditions. J Hazard Mater. 2022;6:100055. https://doi.org/10.1016/j.hazadv.2022.100055.

    Article  CAS  Google Scholar 

  19. Shukla N, Singh D, Tripathi A, Kumari P, Gupta RK, Singh S, Shanker K, Singh A. Synergism of endophytic Bacillus subtilis and Klebsiella aerogenes modulates plant growth and bacoside biosynthesis in Bacopa monnieri. Front Plant Sci. 2022;13:896856. https://doi.org/10.3389/fpls.2022.896856.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Khan AL, Waqas M, Kang SM, Al-Harrasi A, Hussain J, Al-Rawahi A, Lee IJ. Bacterial endophyte Sphingomonas sp. LK11 produces gibberellins and IAA and promotes tomato plant growth. J Microbiol. 2014;52:689–95. https://doi.org/10.1007/s12275-014-4002-7.

    Article  CAS  PubMed  Google Scholar 

  21. Kumar V, Jain L, Jain SK, Chaturvedi S, Kaushal P. Bacterial endophytes of rice (Oryza sativa L.) and their potential for plant growth promotion and antagonistic activities. S Afr J Bot. 2020;134:50–63. https://doi.org/10.1016/j.sajb.2020.02.017.

    Article  CAS  Google Scholar 

  22. Silva Santos SD, Silva AAD, Polonio JC, Polli AD, Orlandelli RC, Oliveira JADS, Pamphile JA. Influence of plant growth-promoting endophytes Colletotrichum siamense and Diaporthe masirevici on tomato plants (Lycopersicon esculentum Mill). Mycology. 2022;13(4):257–70. https://doi.org/10.1080/21501203.2022.2050825.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Shrivastava G, Ownley BH, Augé RM, Toler H, Dee M, Vu A, Chen F. Colonization by arbuscular mycorrhizal and endophytic fungi enhanced terpene production in tomato plants and their defense against a herbivorous insect. Symbiosis. 2015;65:65–74. https://doi.org/10.1007/s13199-015-0319-1.

    Article  CAS  Google Scholar 

  24. Motoyama T. Secondary metabolites of the rice blast fungus Pyricularia oryzae: Biosynthesis and biological function. Int J Mol Sci. 2020;21(22):8698. https://doi.org/10.3390/ijms21228698.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shahzad R, Waqas M, Khan AL, Al-Hosni K, Kang SM, Seo CW, Lee IJ. Indoleacetic acid production and plant growth promoting potential of bacterial endophytes isolated from rice (Oryza sativa L.) seeds. Acta Biol Hung. 2017;68(2):175–86. https://doi.org/10.1556/018.68.2017.2.5.

    Article  CAS  PubMed  Google Scholar 

  26. Bajaj R, Huang Y, Gebrechristos S, Mikolajczyk B, Brown H, Prasad R, Bushley KE. Transcriptional responses of soybean roots to colonization with the root endophytic fungus Piriformospora indica reveals altered phenylpropanoid and secondary metabolism. Sci Rep. 2018;8(1):10227. https://doi.org/10.1007/s41598-018-26809-3.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Nifakos K, Tsalgatidou PC, Thomloudi EE, Skagia A, Kotopoulis D, Baira E, Katinakis P. Genomic analysis and secondary metabolites production of the endophytic Bacillus velezensis Bvel1: a biocontrol agent against Botrytis cinerea causing bunch rot in post-harvest table grapes. Plants. 2021;10(8):1716. https://doi.org/10.3390/plants10081716.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pan SY, Zhou SF, Gao SH, Yu ZL, Zhang SF, Tang MK, Ko KM. New perspectives on how to discover drugs from herbal medicines: CAM’s outstanding contribution to modern therapeutics. Evidence-Based Complem Altern Med. 2013. https://doi.org/10.1155/2013/627375.

    Article  Google Scholar 

  29. Erb M, Kliebenstein DJ. Plant secondary metabolites as defenses, regulators, and primary metabolites: the blurred functional trichotomy. Plant Physiol. 2020;184(1):39–52. https://doi.org/10.1104/pp.20.00433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bino RJ, Hall RD, Fiehn O, Kopka J, Saito K, Draper J, et al. Potential of metabolomics as a functional genomics tool. Trends Plant Sci. 2004;9(9):418–25. https://doi.org/10.1016/j.tplants.2004.07.004.

    Article  CAS  PubMed  Google Scholar 

  31. McMurry JE. Organic chemistry with biological applications. Secondary metabolites: an introduction to natural products chemistry. Stamford: Cengage Learning Ltd.; 2015. p. 1016–46.

    Google Scholar 

  32. Jiménez GS, Ducoing HP, Sosa MR. La participación de los metabolitos secundarios en la defensa de las plantas. Rev Mex Fitopatol. 2003;21(3):355–63. https://doi.org/10.18781/r.mex.fit.0303-7.

    Article  Google Scholar 

  33. Inoue M, Craker LE. Medicinal and aromatic plants-Uses and functions. Horticulture: plants for people and places. J Environ Hortic. 2014;2:645–69. https://doi.org/10.1007/978-94-017-8581-5_3.

    Article  Google Scholar 

  34. Bajaj Y, Furmanowa M, OlszowskA O. Biotechnology of the micropropagation. Med Aromat Plants. 2012;4:60. https://doi.org/10.1007/978-3-642-73026-9_3.

    Article  Google Scholar 

  35. Shafi A, Zahoor I. Metabolomics of medicinal and aromatic plants: goldmines of secondary metabolites for herbal medicine research. In: Aftab T, Hakeem KR, editors. medicinal and aromatic plants. Cambridge: Academic Press; 2021. p. 261–87. https://doi.org/10.1016/B978-0-12-819590-1.00012-4.

    Chapter  Google Scholar 

  36. Calixto JB. The role of natural products in modern drug discovery. Anais da Academia Brasileira de Ciências. 2019. https://doi.org/10.1590/0001-3765201920190105.

    Article  PubMed  Google Scholar 

  37. Farnsworth NR. Studies on Catharanthus alkaloids. VI. Evaluation by means of thin-layer chromatography and ceric ammonium sulfate spray reagent. Lloydia. 1964;27:302–14.

    CAS  Google Scholar 

  38. Abdullahi AD, Mustapha RK, Yau S, Adam MS. Exploring the Nigerian medicinal plants with anticancer activities: a pharmacological review. Mod chem. 2018;6(2):35–8. https://doi.org/10.11648/j.mc.20180602.14.

    Article  CAS  Google Scholar 

  39. Zhou J, Liu Z, Wang S, Li J, Li Y, Chen WK, Wang R. Fungal endophytes promote the accumulation of Amaryllidaceae alkaloids in Lycoris radiata. Environ Microbiol. 2020;22(4):1421–34. https://doi.org/10.1111/1462-2920.14958.

    Article  CAS  PubMed  Google Scholar 

  40. Harrison JG, Griffin EA. The diversity and distribution of endophytes across biomes, plant phylogeny and host tissues: how far have we come and where do we go from here? Environ Microbiol. 2020;22(6):2107–23. https://doi.org/10.1111/1462-2920.14968.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Xiong C, Zhu YG, Wang JT, Singh B, Han LL, Shen JP, He JZ. Host selection shapes crop microbiome assembly and network complexity. New Phytol. 2021;229(2):1091–104. https://doi.org/10.1111/nph.16890.

    Article  CAS  PubMed  Google Scholar 

  42. Singh PK, Egidi E, Macdonald CA, Singh BK. Host selection has a stronger impact on leaf microbiome assembly compared to land-management practices. J Sustain Agric Environ. 2023. https://doi.org/10.1002/sae2.12043.

    Article  Google Scholar 

  43. Kandel SL, Joubert PM, Doty SL. Bacterial endophyte colonization and distribution within plants. Microorganisms. 2017;5(4):77. https://doi.org/10.3390/microorganisms5040077.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Vandenkoornhuyse P, Quaiser A, Duhamel M, Le Van A, Dufresne A. The importance of the microbiome of the plant holobiont. New Phytol. 2015;206(4):1196–206. https://doi.org/10.1111/nph.13312.

    Article  PubMed  Google Scholar 

  45. Pandey SS, Jain R, Bhardwaj P, Thakur A, Kumari M, Bhushan S, Kumar S. Plant probiotics-endophytes pivotal to plant health. Microbiol Res. 2022;263:127148. https://doi.org/10.1016/j.micres.2022.127148.

    Article  CAS  PubMed  Google Scholar 

  46. Purushotham N, Jones E, Monk J, Ridgway H. Community structure, diversity and potential of endophytic bacteria in the primitive New Zealand medicinal plant Pseudowintera colorata. Plants. 2020;9(2):156. https://doi.org/10.3390/plants9020156.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Abdelfattah A, Wisniewski M, Droby S, Schena L. Spatial and compositional variation in the fungal communities of organic and conventionally grown apple fruit at the consumer point-of-purchase. Hort Res. 2016;3:16047. https://doi.org/10.1038/hortres.2016.47.

    Article  CAS  Google Scholar 

  48. Sauer S, Dlugosch L, Kammerer DR, Stintzing FC, Simon M. The microbiome of the medicinal plants Achillea millefolium L. and Hamamelis virginiana L. Front Microbiol. 2021;12:696398. https://doi.org/10.3389/fmicb.2021.696398.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Caruso G, Abdelhamid MT, Kalisz A, Sekara A. Linking endophytic fungi to medicinal plants therapeutic activity. A case study on Asteraceae. Agriculture. 2020;10(7):286. https://doi.org/10.3390/agriculture10070286.

    Article  CAS  Google Scholar 

  50. Hassan SED. Plant growth-promoting activities for bacterial and fungal endophytes isolated from medicinal plant of Teucrium polium L. J Adv Res. 2017;8(6):687–95. https://doi.org/10.1016/j.jare.2017.09.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kumar V, Kumar A, Pandey KD, Roy BK. Isolation and characterization of bacterial endophytes from the roots of Cassia tora L. Ann Microbiol. 2015;65(3):1391–9. https://doi.org/10.1007/s13213-014-0977-x.

    Article  CAS  Google Scholar 

  52. Govarthanan M, Mythili R, Selvankumar T, Kamala-Kannan S, Rajasekar A, Chang YC. Bioremediation of heavy metals using an endophytic bacterium Paenibacillus spRM isolated from the roots of Tridax procumbens. 3 Biotech. 2016;6(2):242. https://doi.org/10.1007/s13205-016-0560-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kumar A, Singh R, Yadav A, Giri DD, Singh PK, Pandey KD. Isolation and characterization of bacterial endophytes of Curcuma longa L. 3 Biotech. 2016;6(1):60. https://doi.org/10.1007/s13205-016-0393-y.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Kumar A, Verma H, Singh VK, Singh PP, Singh SK, Ansari WA, Yadav A, Singh PK, Pandey KD. Role of Pseudomonas sp. in sustainable agriculture and disease management. In: Meena VS, Mishra PK, Bisht JK, Pattanayak A, editors. Agriculturally important microbes for sustainable agriculture. Singapore: Springer; 2017. p. 195–215. https://doi.org/10.1007/978-981-10-5343-6_7.

    Chapter  Google Scholar 

  55. Khan Z, Doty SL. Characterization of bacterial endophytes of sweet potato plants. Plant Soil. 2009;322(1–2):197–207. https://doi.org/10.1007/s11104-009-9908-1.

    Article  CAS  Google Scholar 

  56. Bai Y, D’Aoust F, Smith DL, Driscoll BT. Isolation of plant-growth-promoting Bacillus strains from soybean root nodules. Can J Microbiol. 2002;48(3):230–8. https://doi.org/10.1139/w02-014.

    Article  CAS  PubMed  Google Scholar 

  57. Jasim B, Joseph AA, John CJ, Mathew J, Radhakrishnan EK. Isolation and characterization of plant growth promoting endophytic bacteria from the rhizome of Zingiber officinale. 3 Biotech. 2014;4(2):197–204. https://doi.org/10.1007/s13205-013-0143-3.

    Article  CAS  PubMed  Google Scholar 

  58. Forchetti G, Masciarelli O, Alemano S, Alvarez D, Abdala G. Endophytic bacteria in sunflower (Helianthus annuus L.): isolation, characterization, and production of jasmonates and abscisic acid in culture medium. Appl Microbiol Biotechnol. 2007;76(5):1145–52. https://doi.org/10.1007/s00253-007-1077-7.

    Article  CAS  PubMed  Google Scholar 

  59. Dias AC, Costa FE, Andreote FD, Lacava PT, Teixeira MA, Assumpçao LC, Araújo WL, Azevedo JL, Melo IS. Isolation of micropropagated strawberry endophytic bacteria and assessment of their potential for plant growth promotion. World J Microbiol Biotechnol. 2009;25(2):189–95. https://doi.org/10.1007/s11274-008-9878-0.

    Article  CAS  Google Scholar 

  60. Tiwari R, Kalra A, Darokar MP, Chandra M, Aggarwal N, Singh AK, Khanuja SPS. Endophytic bacteria from Ocimum sanctum and their yield enhancing capabilities. Curr Microbiol. 2010;60(3):167–71. https://doi.org/10.1007/s00284-009-9520-x.

    Article  CAS  PubMed  Google Scholar 

  61. Yaish MW, Antony I, Glick BR. Isolation and characterization of endophytic plant growth-promoting bacteria from date palm tree (Phoenix dactylifera L) and their potential role in salinity tolerance. Antonie Van Leeuwenhoek. 2015;107(6):1519–32. https://doi.org/10.1007/s10482-015-0445-z.

    Article  CAS  PubMed  Google Scholar 

  62. Bhore SJ, Ravichantar N, Loh CY. Screening of endophytic bacteria isolated from leaves of Sambung nyawa [Gynura procumbens (Lour) Merr.] for cytokinin-like compounds. Bioinform. 2010;5(5):191. https://doi.org/10.6026/97320630005191.

    Article  Google Scholar 

  63. Sun K, Liu J, Gao Y, Jin L, Gu Y, Wang W. Isolation, plant colonization potential, and phenanthrene degradation performance of the endophytic bacterium Pseudomonas sp Ph6-gfp. Sci Rep. 2014;4:5462. https://doi.org/10.1038/srep05462.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wang XM, Yang B, Ren CG, Wang HW, Wang JY, Dai CC. Involvement of abscisic acid and salicylic acid in signal cascade regulating bacterial endophyte-induced volatile oil biosynthesis in plantlets of Atractylodes lancea. Physiol Plantarum. 2015;153(1):30–42. https://doi.org/10.1111/ppl.12236.

    Article  CAS  Google Scholar 

  65. Babu AG, Kim JD, Oh BT. Enhancement of heavy metal phytoremediation by Alnus firma with endophytic Bacillus thuringiensis GDB-1. J Hazard Mater. 2013;250:477–83. https://doi.org/10.1016/j.jhazmat.2013.02.014.

    Article  CAS  PubMed  Google Scholar 

  66. Xu M, Sheng J, Chen L, Men Y, Gan L, Guo S, Shen L. Bacterial community compositions of tomato (Lycopersicum esculentum Mill) seeds and plant growth promoting activity of ACC deaminase producing Bacillus subtilis (HYT-12-1) on tomato seedlings. World J Microbiol Biotechnol. 2014;30(3):835–45. https://doi.org/10.1007/s11274-013-1486-y.

    Article  CAS  PubMed  Google Scholar 

  67. Karthikeyan B, Joe MM, Islam MR, Sa T. ACC deaminase containing diazotrophic endophytic bacteria ameliorate salt stress in Catharanthus roseus through reduced ethylene levels and induction of antioxidative defense systems. Symbiosis. 2012;56:77–86. https://doi.org/10.1007/s13199-012-0162-6.

    Article  CAS  Google Scholar 

  68. El-Deeb B, Fayez K, Gherbawy Y. Isolation and characterization of endophytic bacteria from Plectranthus tenuiflorus medicinal plant in Saudi Arabia desert and their antimicrobial activities. J Plant Interact. 2013;8(1):56–64. https://doi.org/10.1080/17429145.2012.680077.

    Article  CAS  Google Scholar 

  69. Sun L, Lu Z, Bie X, Lu F, Yang S. Isolation and characterization of a co-producer of fengycins and surfactins, endophytic Bacillus amyloliquefaciens ES-2, from Scutellaria baicalensis Georgi. World J Microbiol Biotechnol. 2006;22(12):1259–66. https://doi.org/10.1007/s11274-006-9170-0.

    Article  CAS  Google Scholar 

  70. Chen L, Luo S, Xiao X, Guo H, Chen J, Wan Y, Li B, Xu T, Xi Q, Rao C, Liu C. Application of plant growth-promoting endophytes (PGPE) isolated from Solanum nigrum L. for phytoextraction of Cd-polluted soils. Appl Soil Ecol. 2010;46(3):383–9. https://doi.org/10.1016/j.apsoil.2010.10.003.

    Article  Google Scholar 

  71. Araújo WL, Maccheroni W, Azevedo JL. Characterization of an endophytic bacterial community associated with Eucalyptus spp. Genet Mol Res. 2009;8(4):1408–22. https://doi.org/10.4238/vol8-4gmr691.

    Article  PubMed  Google Scholar 

  72. Pandey SS, Singh S, Babu CS, Shanker K, Srivastava NK, Kalra A. Endophytes of Opium poppy differentially modulate host plant productivity and genes for the biosynthetic pathway of benzylisoquinoline alkaloids. Planta. 2016;243(5):1097–114. https://doi.org/10.1007/s00425-016-2467-9.

    Article  CAS  PubMed  Google Scholar 

  73. Abdallah RB, Mejdoub-Trabelsi B, Nefzi A, Jabnoun-Khiareddine H, Daami-Remadi M. Isolation of endophytic bacteria from Withania somnifera and assessment of their ability to suppress Fusarium wilt disease in tomato and to promote plant growth. J Plant Pathol Microbiol. 2016;7(5):2–11. https://doi.org/10.4172/2157-7471.1000352.

    Article  CAS  Google Scholar 

  74. Latif Khan A, Ahmed Halo B, Elyassi A, Ali S, Al-Hosni K, Hussain J, Al-Harrasi A, Lee IJ. Indole acetic acid and ACC deaminase from endophytic bacteria improves the growth of Solarium lycopersicum. Electron J Biotechnol. 2016;19(3):58–64. https://doi.org/10.1016/j.ejbt.2016.02.001.

    Article  Google Scholar 

  75. Trivedi P, Spann T, Wang N. Isolation and characterization of beneficial bacteria associated with citrus roots in Florida. Microb Ecol. 2011;62(2):324–36. https://doi.org/10.1007/s00248-011-9822-y.

    Article  CAS  PubMed  Google Scholar 

  76. Sunkar S, Akshaya A, Aarthi B, Nachiyar VC, Prakash P. Phytochemical analysis and isolation of endophytic bacteria from Bauhinia purpurea. Res J Pharm Technol. 2018;11(5):1867–76. https://doi.org/10.5958/0974-360X.2018.00347.5.

    Article  Google Scholar 

  77. Joe MM, Devaraj S, Benson A, Sa T. Isolation of phosphate solubilizing endophytic bacteria from Phyllanthus amarus Schum & Thonn: evaluation of plant growth promotion and antioxidant activity under salt stress. J Appl Res Med Aromat Plants. 2016;3(2):71–7. https://doi.org/10.1016/j.jarmap.2016.02.003.

    Article  Google Scholar 

  78. Aung TN, Nourmohammadi S, Sunitha EM, Myint M. Isolation of endophytic bacteria from green gram and study on their plant growth promoting activities. Intl J Appl Biol Pharmacol Tech. 2011;2:525–36.

    Google Scholar 

  79. Sona Janarthine SR, Eganathan P, Balasubramanian T, Vijayalakshmi S. Endophytic bacteria isolated from the pneumatophores of Avicennia marina. Afr J Microbiol Res. 2011;5(26):4455–66. https://doi.org/10.5897/AJMR10.188.

    Article  CAS  Google Scholar 

  80. Sheng XF, Xia JJ, Jiang CY, He LY, Qian M. Characterization of heavy metal resistant endophytic bacteria from rape (Brassica napus) roots and their potential in promoting the growth and lead accumulation of rape. Environ Pollut. 2008;156(3):1164–70. https://doi.org/10.1016/j.envpol.2008.04.007.

    Article  CAS  PubMed  Google Scholar 

  81. Sessitsch A, Coenye T, Sturz AV, Vandamme P, Barka EA, Salles JF, Van Elsas JD, Faure D, Reiter B, Glick BR, Wang-Pruski G, Nowak J. Burkholderia phytofirmans sp. Nov., a novel plant-associated bacterium with plant-beneficial properties. Int J Syst Evol. 2005;55(3):1187–92. https://doi.org/10.1099/ijs.0.63149-0.

    Article  CAS  Google Scholar 

  82. Dong Z, Canny MJ, McCully ME, Roboredo MR, Cabadilla CF, Ortega E, Rodes R. A nitrogen-fixing endophyte of sugarcane stems (a new role for the apoplast). Plant Physiol. 1994;105(4):1139–47. https://doi.org/10.1104/pp.105.4.1139.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Ji X, Lu G, Gai Y, Zheng C, Mu Z. Biological control against bacterial wilt and colonization of mulberry by an endophytic Bacillus subtilis strain. FEMS Microbiol Ecol. 2008;65(3):565–73. https://doi.org/10.1111/j.1574-6941.2008.00543.x.

    Article  CAS  PubMed  Google Scholar 

  84. Riggs PJ, Chelius MK, Iniguez AL, Kaeppler SM, Triplett EW. Enhanced maize productivity by inoculation with diazotrophic bacteria. Aust J Plant Physiol. 2001;28:829–36. https://doi.org/10.1071/PP01045.

    Article  Google Scholar 

  85. Liu X, Zhao H, Chen S. Colonization of maize and rice plants by strain Bacillus megaterium C4. Curr Microbiol. 2006;52(3):186–90. https://doi.org/10.1007/s00284-005-0162-3.

    Article  CAS  PubMed  Google Scholar 

  86. Govindarajan M, Balandreau J, Muthukumarasamy R, Revathi G, Lakshminarasimhan C. Improved yield of micropropagated sugarcane following inoculation by endophytic Burkholderia vietnamiensis. Plant Soil. 2006;280:239–52. https://doi.org/10.1007/s11104-005-3223-2.

    Article  CAS  Google Scholar 

  87. Germaine KJ, Liu X, Cabellos GG, Hogan JP, Ryan D, Dowling DN. Bacterial endophyte-enhanced phytoremediation of the organochlorine herbicide 2, 4-dichlorophenoxyacetic acid. FEMS Microbiol Ecol. 2006;57(2):302–10. https://doi.org/10.1111/j.1574-6941.2006.00121.x.

    Article  CAS  PubMed  Google Scholar 

  88. Oliveira ALM, Stoffels M, Schmid M, Reis VM, Baldani JI. Colonization of sugarcane plantlets by mixed inoculations with diazotrophic bacteria. Eur J Soil Biol. 2009;45:106–13. https://doi.org/10.1111/j.1472-765X.2010.02899.x.

    Article  CAS  Google Scholar 

  89. Oliveira ALM, Urquiaga S, Döbereiner J, Baldani JI. The effect of inoculating endophytic N2-fixing bacteria on micropropagated sugarcane plants. Plant Soil. 2002;242:205–15. https://doi.org/10.1023/A:1016249704336.

    Article  CAS  Google Scholar 

  90. Chen L, Luo S, Chen J, Wan Y, Liu C, Liu Y, Zeng G. Diversity of endophytic bacterial populations associated with Cd-hyperaccumulator plant Solanum nigrum L. grown in mine tailings. Appl Soil Ecol. 2012;62:24–30. https://doi.org/10.1016/j.apsoil.2012.07.005.

    Article  Google Scholar 

  91. Nayak BK. Enumeration of phylloplane and endophytic fungi from medicinal plant, Solanum nigrum by two different techniques. Int J Chem Concept. 2015;1(3):103–8. https://doi.org/10.2478/ijcc20150018.

    Article  Google Scholar 

  92. El-Hawary SS, Mohammed R, AbouZid SF, Bakeer W, Ebel R, Sayed AM, Rateb ME. Solamargine production by a fungal endophyte of Solanum nigrum. J Appl Microbiol. 2016;120(4):900–11. https://doi.org/10.1111/jam.13077.

    Article  CAS  PubMed  Google Scholar 

  93. Abdallah RAB, Jabnoun-Khiareddine H, Nefzi A, Ayed F, Daami-Remadi M. Endophytic bacteria from Solanum nigrum with plant growth-promoting and Fusarium wilt-suppressive abilities in tomato. Tun J Plant Protec. 2018;13:157–82.

    Google Scholar 

  94. Chowdhury DR, Chatterjee SK, Roy SK. Studies on endophytic fungi of Calotropis procera (L.) R. Br. with a view to their antimicrobial and antioxidant actvities mediated by extracellular synthesised silver nanoparticles. IOSR J Pharm Biol Sci. 2016;11(5):113–21. https://doi.org/10.9790/3008-110502113121.

    Article  Google Scholar 

  95. Selvanathan S, Indrakumar I, Johnpaul M. Biodiversity of the endophytic fungi isolated from Calotropis gigantea (L.) R. Br. Recent res sci technol. 2011. https://doi.org/10.4314/rrst.v3i4.6.

    Article  Google Scholar 

  96. Tavarideh F, Pourahmad F, Nemati M. Diversity and antibacterial activity of endophytic bacteria associated with medicinal plant, Scrophularia striata. Vet Res Forum. 2022;13(3):409. https://doi.org/10.30466/vrf.2021.529714.3174.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Akinsanya MA, Goh JK, Lim SP, Ting ASY. Metagenomics study of endophytic bacteria in Aloe vera using next-generation technology. Genom Data. 2015;6:159–63. https://doi.org/10.1016/j.gdata.2015.09.004.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Aravind R, Kumar A, Eapen SJ, Ramana KV. Endophytic bacterial flora in root and stem tissues of black pepper (Piper nigrum L.) genotype: isolation, identification and evaluation against Phytophthora capsici. Lett Appl Microbiol. 2009;48(1):58–64. https://doi.org/10.1111/j.1472-765X.2008.02486.x.

    Article  CAS  PubMed  Google Scholar 

  99. Gagne S, Richard H, Rousseau H, Antoun H. Xylem residing bacteria in alfalfa roots. Can J Microbiol. 1987;33:996–1000. https://doi.org/10.1139/m87-175.

    Article  Google Scholar 

  100. Stajkovic O, De Meyer S, Milicic B, Willems A, Delic D. Isolation and characterization of endophytic nonrhizobial bacteria from root nodules of alfalfa (Medicago sativa L.). Bot Serb. 2009;33(1):107–14.

    Google Scholar 

  101. Ibrahim E, Fouad H, Zhang M, Zhang Y, Qiu W, Yan C, Li B, Mo J, Chen J. Biosynthesis of silver nanoparticles using endophytic bacteria and their role in inhibition of rice pathogenic bacteria and plant growth promotion. RSC Adv. 2019;9(50):29293–9. https://doi.org/10.1039/C9RA04246F.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Vendan RT, Yu YJ, Lee SH, Rhee YH. Diversity of endophytic bacteria in ginseng and their potential for plant growth promotion. J Microbiol Res. 2010;48:559–65. https://doi.org/10.1007/s12275-010-0082-1.

    Article  CAS  Google Scholar 

  103. Aswathy AJ, Jasim B, Jyothis M, Radhakrishnan EK. Identification of two strains of Paenibacillus sp. as indole 3 acetic acid producing rhizome-associated endophytic bacteria from Curcuma longa. 3 Biotech. 2013;3:219–24. https://doi.org/10.1007/s13205-012-0086-0.

    Article  PubMed  Google Scholar 

  104. Jayakumar A, Krishna A, Mohan M, Nair IC, Radhakrishnan EK. Plant growth enhancement, disease resistance, and elemental modulatory effects of plant probiotic endophytic Bacillus sp. Fcl1. Probiotics Antimicrob Proteins. 2019;11(2):526–34. https://doi.org/10.1007/s12602-018-9417-8.

    Article  CAS  PubMed  Google Scholar 

  105. Nxumalo CI, Ngidi LS, Shandu JSE, Maliehe TS. Isolation of endophytic bacteria from the leaves of Anredera cordifolia CIX1 for metabolites and their biological activities. BMC Complement Med Ther. 2020;20(1):300. https://doi.org/10.1186/s12906-020-03095-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Cardoso VM, Campos FF, Santos ARO, Ottoni MHF, Rosa CA, Almeida VG, Grael CFF. Biotechnological applications of the medicinal plant Pseudobrickellia brasiliensis and its isolated endophytic bacteria. J Appl Microbiol. 2020;129(4):926–34. https://doi.org/10.1038/nature11336.

    Article  CAS  PubMed  Google Scholar 

  107. Devi KA, Pandey G, Rawat AKS, Sharma GD, Pandey P. The endophytic symbiont-Pseudomonas aeruginosa stimulates the antioxidant activity and growth of Achyranthes aspera L. Front Microbiol. 2017;8:1897. https://doi.org/10.3389/fmicb.2017.01897.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Karpinets TV, Park BH, Syed MH, Klotz MG, Uberbacher EC. Metabolic environments and genomic features associated with pathogenic and mutualistic interactions between bacteria and plants. Mol Plant Microbe Interact. 2014;27(7):664–77. https://doi.org/10.1094/MPMI-12-13-0368-R.

    Article  CAS  PubMed  Google Scholar 

  109. Compant S, Clément C, Sessitsch A. Plant growth-promoting bacteria in the rhizo-and endosphere of plants: their role, colonization, mechanisms involved and prospects for utilization. Soil Biol Biochem. 2010;42(5):669–78. https://doi.org/10.1016/j.soilbio.2009.11.024.

    Article  CAS  Google Scholar 

  110. Wu L, Han T, Li W, Jia M, Xue L, Rahman K, Qin L. Geographic and tissue influences on endophytic fungal communities of Taxus chinensis var. mairei in China. Curr Microbiol. 2013;66(1):40–8. https://doi.org/10.1007/s00284-012-0235-z.

    Article  CAS  PubMed  Google Scholar 

  111. Chi F, Shen SH, Cheng HP, Jing YX, Yanni YG, Dazzo FB. Ascending migration of endophytic rhizobia, from roots to leaves, inside rice plants and assessment of benefits to rice growth physiology. Appl Environ Microbiol. 2005;71(11):7271–8. https://doi.org/10.1128/AEM.71.11.7271-7278.2005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Rosenblueth M, Martínez-Romero E. Bacterial endophytes and their interactions with hosts. Mol Plant Microbe Interact. 2006;19(8):827–37. https://doi.org/10.1002/jobm.202000657.

    Article  CAS  PubMed  Google Scholar 

  113. Brader G, Compant S, Mitter B, Trognitz F, Sessitsch A. Metabolic potential of endophytic bacteria. Curr Opin Biotechnol. 2014;27:30–7. https://doi.org/10.1016/j.copbio.2013.09.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Aeron A, Khare E, Jha CK, Meena VS, Aziz SMA, Islam MT, Meena RK. Revisiting the plant growth-promoting rhizobacteria: lessons from the past and objectives for the future. Arch Microbiol. 2020;2020(202):665–76. https://doi.org/10.1007/s00203-019-01779-w.

    Article  CAS  Google Scholar 

  115. Ma L, Wang WQ, Shi R, Zhang XM, Li X, Yang YS, Mo MH. Effects of organic acids on the chemotaxis profiles and biocontrol traits of antagonistic bacterial endophytes against root-rot disease in Panax notoginseng. Antonie Van Leeuwenhoek. 2021;114:1771–89. https://doi.org/10.1007/s10482-021-01636-1.

    Article  CAS  PubMed  Google Scholar 

  116. Sauer K, Camper AK. Characterization of phenotypic changes in Pseudomonas putida in response to surface-associated growth. J Bacteriol. 2001;183(22):6579–89. https://doi.org/10.1128/JB.183.22.6579-6589.2001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Lahrmann U, Zuccaro A. Opprimo ergo sum—evasion and suppression in the root endophytic fungus Piriformospora indica. Mol Plant Microbe Interact. 2012;25(6):727–37. https://doi.org/10.1094/MPMI-11-11-0291.

    Article  CAS  PubMed  Google Scholar 

  118. Zinniel DK, Lambrecht P, Harris NB, Feng Z, Kuczmarski D, Higley P, Ishimaru CA, Arunakumari A, Barletta RG, Vidaver AK. Isolation and characterization of endophytic colonizing bacteria from agronomic crops and prairie plants. Appl Environ Microbiol. 2002;68(5):2198–208. https://doi.org/10.1128/AEM.68.5.2198-2208.2002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Hardoim PR, van Overbeek LS, van Elsas JD. Properties of bacterial endophytes and their proposed role in plant growth. Trends Microbiol. 2008;16(10):463–71. https://doi.org/10.1016/j.tim.2008.07.008.

    Article  CAS  PubMed  Google Scholar 

  120. Reinhold-Hurek B, Maes T, Gemmer S, Van Montagu M, Hurek T. An endoglucanase is involved in infection of rice roots by the not-cellulose-metabolizing endophyte Azoarcus sp. strain BH72. Mol Plant Microbe Interact. 2006;19:181–8. https://doi.org/10.1094/MPMI-19-0181.

    Article  CAS  PubMed  Google Scholar 

  121. Naveed M, Mitter B, Yousaf S, Pastar M, Afzal M, Sessitsch A. The endophyte Enterobacter sp FD17: a maize growth enhancer selected based on rigorous testing of plant beneficial traits and colonization characteristics. Biol Fertil Soils. 2014;2014(50):249–62. https://doi.org/10.1007/s00374-013-0854-y.

    Article  CAS  Google Scholar 

  122. Gasser I, Cardinale M, Müller H, Heller S, Eberl L, Lindenkamp N, Kaddor C, Steinbüchel A, Berg G. Analysis of the endophytic lifestyle and plant growth promotion of Burkholderia terricola ZR2-12. Plant Soil. 2011;347(1–2):125. https://doi.org/10.1007/s11104-011-0833-8.

    Article  CAS  Google Scholar 

  123. Krause A, Ramakumar A, Bartels D, Battistoni F, Bekel T, Boch J, Böhm M, Friedrich F, Hurek T, Krause L, Linke B. Complete genome of the mutualistic N2-fixing grass endophyte Azoarcus sp. strain BH72. Nat Biotechnol. 2006;24(11):1384–91. https://doi.org/10.1038/nbt1243.

    Article  CAS  Google Scholar 

  124. Getachew G, Rewald B, Godbold DL, Sandén H. Endophytic fungal root colonization of Eragrostis tef in eroded croplands of the Ethiopian highlands is limited by low spore density and fertilisation. Agronomy. 2019;9(2):73. https://doi.org/10.3390/agronomy9020073.

    Article  CAS  Google Scholar 

  125. Nizam S, Qiang X, Wawra S, Nostadt R, Getzke F, Schwanke F, Dreyer I, Langen G, Zuccaro A. Serendipita indica E5’NT modulates extracellular nucleotide levels in the plant apoplast and affects fungal colonization. EMBO Rep. 2019;20(2):e47430. https://doi.org/10.15252/embr.201847430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Liu H, Brettell LE, Qiu Z, Singh BK. Microbiome-mediated stress resistance in plants. Trends Plant Sci. 2020;25(8):733–43. https://doi.org/10.1016/j.tplants.2020.03.014.

    Article  CAS  PubMed  Google Scholar 

  127. Trivedi P, Batista BD, Bazany KE, Singh BK. Plant–microbiome interactions under a changing world: responses, consequences and perspectives. New Phytol. 2022;234(6):1951–9. https://doi.org/10.1111/nph.18016.

    Article  PubMed  Google Scholar 

  128. Singh BK, Delgado-Baquerizo M, Egidi E, Guirado E, Leach JE, Liu H, Trivedi P. Climate change impacts on plant pathogens, food security and paths forward. Nat Rev Microbiol. 2023. https://doi.org/10.1038/s41579-023-00900-7.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Fu Y, Yin ZH, Yin CY. Biotransformation of ginsenoside Rb1 to ginsenoside Rg3 by endophytic bacterium Burkholderia sp. GE 17–7 isolated from Panax ginseng. J Appl Microbiol. 2017;122(6):1579–85. https://doi.org/10.1111/jam.13435.

    Article  CAS  PubMed  Google Scholar 

  130. Ye B, Wu Y, Zhai X, Zhang R, Wu J, Zhang C, Zheng C. Beneficial effects of endophytic fungi from the Anoectochilus and Ludisia species on the growth and secondary metabolism of Anoectochilus roxburghii. ACS Omega. 2020;5(7):3487–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Salehi M, Farhadi S, Moieni A, Safaie N, Ahmadi H. Mathematical modeling of growth and paclitaxel biosynthesis in Corylus avellana cell culture responding to fungal elicitors using multilayer perceptron-genetic algorithm. Front Plant Sci. 2020;11:1148. https://doi.org/10.3389/fpls.2020.01148.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Singh S, Pandey SS, Tiwari R, Pandey A, Shanker K, Kalra A. Endophytic consortium with growth-promoting and alkaloid enhancing capabilities enhance key terpenoid indole alkaloids of Catharanthus roseus in the winter and summer seasons. Ind Crops Prod. 2021;166:113437. https://doi.org/10.1016/j.indcrop.2021.113437.

    Article  CAS  Google Scholar 

  133. Singh D, Thapa S, Mahawar H, Kumar D, Geat N, Singh SK. Prospecting potential of endophytes for modulation of biosynthesis of therapeutic bioactive secondary metabolites and plant growth promotion of medicinal and aromatic plants. Antonie Van Leeuwenhoek. 2022;115(6):699–730. https://doi.org/10.1007/s10482-022-01736-6.

    Article  CAS  PubMed  Google Scholar 

  134. Chaparro JM, Badri DV, Vivanco JM. Rhizosphere microbiome assemblage is affected by plant development. ISME J. 2014;8(4):790–803. https://doi.org/10.1038/ismej.2013.196.

    Article  CAS  PubMed  Google Scholar 

  135. Sasse J, Martinoia E, Northen T. Feed your friends: do plant exudates shape the root microbiome? Trends Plant Sci. 2018;23(1):25–41. https://doi.org/10.1016/j.tplants.2017.09.003.

    Article  CAS  PubMed  Google Scholar 

  136. Patchett A, Newman JA. Comparison of plant metabolites in root exudates of Lolium perenne infected with different strains of the fungal endophyte Epichloë festucae var. lolii. J Fungus. 2021;7(2):148. https://doi.org/10.3390/jof7020148.

    Article  CAS  Google Scholar 

  137. Fan Y, Gao L, Chang P, Li Z. Endophytic fungal community in grape is correlated to foliar age and domestication. Ann Microbiol. 2020;70:1–8. https://doi.org/10.1186/s13213-020-01574-9.

    Article  Google Scholar 

  138. Wei G, Chen Z, Wang B, Wei F, Zhang G, Wang Y, Zhu G, Zhou Y, Zhao Q, He M, Dong L. Endophytes isolated from Panax notoginseng converted ginsenosides. Microb Biotechnol. 2021;14(4):1730–46. https://doi.org/10.1111/1751-7915.13842.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Li Z, Xiong K, Wen W, Li L, Xu D. Functional endophytes regulating plant secondary metabolism: current status, prospects and applications. Int J Mol Sci. 2023;24(2):1153. https://doi.org/10.3390/ijms24021153.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Zhang Y, Chen R, Zhang D, Qi S, Liu Y. Metabolite interactions between host and microbiota during health and disease: which feeds the other? Biomed Pharmacother. 2023;160:114295. https://doi.org/10.1016/j.biopha.2023.114295.

    Article  CAS  PubMed  Google Scholar 

  141. Pang Z, Chen J, Wang T, Gao C, Li Z, Guo L, Xu J, Cheng Y. Linking plant secondary metabolites and plant microbiomes: a review. Front Plant Sci. 2021;12:621276. https://doi.org/10.3389/fpls.2021.621276.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Matsumoto A, Takahashi Y. Endophytic actinomycetes: promising source of novel bioactive compounds. J Antibiot. 2017;70(5):514–9. https://doi.org/10.1038/ja.2017.20.

    Article  CAS  Google Scholar 

  143. Pratiwi DR, Sari PT, Ratnadewi D. Cinchona cells performance in in vitro culture: quinine alkaloid production with application of different elicitors. IOP Conf Ser Environ Earth Sci. 2018;185(1):012029. https://doi.org/10.1088/1755-1315/185/1/012029.

    Article  Google Scholar 

  144. Pandey SS, Singh S, Babu CV, Shanker K, Srivastava NK, Shukla AK, Kalra A. Fungal endophytes of Catharanthus roseus enhance vindoline content by modulating structural and regulatory genes related to terpenoid indole alkaloid biosynthesis. Sci Rep. 2016;6(1):26583. https://doi.org/10.1038/srep26583.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Ming Q, Su C, Zheng C, Jia M, Zhang Q, Zhang H, Qin L. Elicitors from the endophytic fungus Trichoderma atroviride promote Salvia miltiorrhiza hairy root growth and tanshinone biosynthesis. J Exp Bot. 2013;64(18):5687–94. https://doi.org/10.1093/jxb/ert342.

    Article  CAS  PubMed  Google Scholar 

  146. Dupont PY, Eaton CJ, Wargent JJ, Fechtner S, Solomon P, Schmid J, Cox MP. Fungal endophyte infection of ryegrass reprograms host metabolism and alters development. New Phytol. 2015;208(4):1227–40. https://doi.org/10.1111/nph.13614.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Mastan A, Bharadwaj RKB, Kushwaha RK, Vivek Babu CS. Functional fungal endophytes in Coleus forskohlii regulate labdane diterpene biosynthesis for elevated forskolin accumulation in roots. Microb Ecol. 2019;78(4):914–26. https://doi.org/10.1007/s00248-019-01376-w.

    Article  CAS  PubMed  Google Scholar 

  148. Mastan A, Rane D, Dastager SG, Babu CV. Molecular insights of fungal endophyte co-inoculation with Trichoderma viride for the augmentation of forskolin biosynthesis in Coleus forskohlii. Phytochem. 2021;184:112654. https://doi.org/10.1016/j.phytochem.2021.112654.

    Article  CAS  Google Scholar 

  149. Chhipa H, Kaushik N. Fungal and bacterial diversity isolated from Aquilaria malaccensis tree and soil, induces agarospirol formation within 3 months after artificial infection. Front Microbiol. 2017;8:1286. https://doi.org/10.3389/fmicb.2017.01286.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Yang HR, Yuan J, Liu LH, Zhang W, Chen F, Dai CC. Endophytic Pseudomonas fluorescens induced sesquiterpenoid accumulation mediated by gibberellic acid and jasmonic acid in Atractylodes macrocephala Koidz plantlets. Plant Cell Tissue Organ Cult. 2019;138:445–57. https://doi.org/10.1007/s11240-019-01640-4.

    Article  CAS  Google Scholar 

  151. Del Giudice L, Massardo DR, Pontieri P, Bertea CM, Mombello D, Carata E, Alifano P. The microbial community of Vetiver root and its involvement into essential oil biogenesis. Environ Microbiol. 2008;10(10):2824–41. https://doi.org/10.1111/j.1462-2920.2008.01703.x.

    Article  CAS  PubMed  Google Scholar 

  152. Gao Y, Liu Q, Zang P, Li X, Ji Q, He Z, Zhang L. An endophytic bacterium isolated from Panax ginseng CA Meyer enhances growth, reduces morbidity, and stimulates ginsenoside biosynthesis. Phytochem Lett. 2015;11:132–8. https://doi.org/10.1016/j.phytol.2014.12.007.

    Article  CAS  Google Scholar 

  153. Zhang FS, Lv YL, Zhao Y, Guo SX. Promoting role of an endophyte on the growth and contents of kinsenosides and flavonoids of Anoectochilus formosanus Hayata, a rare and threatened medicinal Orchidaceae plant. J Zhejiang Univ Sci B. 2013;14:785–92. https://doi.org/10.1631/jzus.B1300056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Gupta S, Chaturvedi P. Enhancing secondary metabolite production in medicinal plants using endophytic elicitors: a case study of Centella asiatica (Apiaceae) and asiaticoside. Endophytes Grow world. 2019. https://doi.org/10.1017/9781108607667.015.

    Article  Google Scholar 

  155. Jiang DF, Ma P, Yang J, Wang X, Xu K, Huang Y, Chen S. Formation of blood resin in abiotic Dracaena cochinchinensis inoculated with Fusarium 9568D. Ying Yong Sheng tai xue bao. J Appl Ecol. 2003;14(3):477–8.

    CAS  Google Scholar 

  156. Prasad R, Garg AP, Varma A. Interaction of medicinal plant with plant growth promoting rhizobacteria and symbiotic fungi. In: Podila G, Varma A, editors. Basic research and applications: mycorrhizae. Microbiology series. Kanpur: IK International, India; 2004. p. 363–407.

    Google Scholar 

  157. Yuan ZL, Dai CC, Li X, Tian LS, Wang XX. Extensive host range of an endophytic fungus affects the growth and physiological functions in rice (Oryza sativa L.). Symbiosis. 2007;43:21–8. https://doi.org/10.1007/BF03161613.

    Article  Google Scholar 

  158. Rai M, Acharya D, Singh A, Varma A. Positive growth responses of the medicinal plants Spilanthes calva and Withania somnifera to inoculation by Piriformospora indica in a field trial. Mycorrhiza. 2001;11:123–8. https://doi.org/10.1007/s005720100115.

    Article  PubMed  Google Scholar 

  159. Baldi A, Jain A, Gupta N, Srivastava AK, Bisaria VS. Co-culture of arbuscular mycorrhiza-like fungi (Piriformospora indica and Sebacina vermifera) with plant cells of Linum album for enhanced production of podophyllotoxins: a first report. Biotechnol Lett. 2008;30:1671–7. https://doi.org/10.1007/s10529-008-9736-z.

    Article  CAS  PubMed  Google Scholar 

  160. Li YC, Tao WY, Cheng L. Paclitaxel production using co-culture of Taxus suspension cells and paclitaxel-producing endophytic fungi in a co-bioreactor. Appl Microbiol Biotechnol. 2009;83:233–9. https://doi.org/10.1007/s00253-009-1856-4.

    Article  CAS  PubMed  Google Scholar 

  161. Li YC, Tao WY. Effects of paclitaxel-producing fungal endophytes on growth and paclitaxel formation of Taxus cuspidata cells. Plant Growth Regul. 2009;58:97–105. https://doi.org/10.1007/s10725-008-9355-7.

    Article  CAS  Google Scholar 

  162. Tang Z, Rao L, Peng G, Zhou M, Shi G, Liang Y. Effects of endophytic fungus and its elicitors on cell status and alkaloid synthesis in cell suspension cultures of Catharanthus roseus. J Med Plants Res. 2011;5(11):2192. https://doi.org/10.5897/JMPR.9000520.

    Article  Google Scholar 

  163. Bajaj R, Agarwal A, Rajpal K, Asthana S, Kumar R, Prasad R, Varma A. Co-cultivation of Curcuma longa with Piriformospora indica enhances the yield and active ingredients. Am J Curr Microbiol. 2014;2(1):6–17.

    Google Scholar 

  164. Ding CH, Wang QB, Guo S, Wang ZY. The improvement of bioactive secondary metabolites accumulation in Rumex gmelini Turcz through co-culture with endophytic fungi. Braz J Microbiol. 2018;49(2):362–9. https://doi.org/10.1016/j.bjm.2017.04.013.

    Article  CAS  PubMed  Google Scholar 

  165. Cui JL, Wang YN, Jiao J, Gong Y, Wang JH, Wang ML. Fungal endophyte-induced salidroside and tyrosol biosynthesis combined with signal cross-talk and the mechanism of enzyme gene expression in Rhodiola crenulata. Sci Rep. 2017;7(1):12540. https://doi.org/10.1038/s41598-017-12895-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Zubek S, Mielcarek S, Turnau K. Hypericin and pseudohypericin concentrations of a valuable medicinal plant Hypericum perforatum L. are enhanced by arbuscular mycorrhizal fungi. Mycorrhiza. 2012;22(2):149–56. https://doi.org/10.1007/s00572-011-0391-1.

    Article  CAS  PubMed  Google Scholar 

  167. Ahlawat S, Saxena P, Ali A, Khan S, Abdin MZ. Comparative study of withanolide production and the related transcriptional responses of biosynthetic genes in fungi elicited cell suspension culture of Withania somnifera in shake flask and bioreactor. Plant Physiol Biochem. 2017;114:19–28. https://doi.org/10.1016/j.plaphy.2017.02.013.

    Article  CAS  PubMed  Google Scholar 

  168. Tripathi A, Awasthi A, Singh S, Sah K, Maji D, Patel VK, Verma RK, Kalra A. Enhancing artemisinin yields through an ecologically functional community of endophytes in Artemisia annua. Ind Crops Prod. 2020;150:112375. https://doi.org/10.1016/j.indcrop.2020.112375.

    Article  CAS  Google Scholar 

  169. Yuan J, Zhang W, Sun K, Tang MJ, Chen PX, Li X, Dai CC. Comparative transcriptomics and proteomics of Atractylodes lancea in response to endophytic fungus Gilmaniella sp. AL12 reveals regulation in plant metabolism. Front Microbiol. 2019;10:1208. https://doi.org/10.3389/fmicb.2019.01208.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Ye B, Wu Y, Zhai X, Zhang R, Wu J, Zhang C, Rahman K, Qin L, Han T, Zheng C. Beneficial effects of endophytic fungi from the Anoectochilus and Ludisia species on the growth and secondary metabolism of Anoectochilus roxburghii. Acs Omega. 2020;5(7):3487–97. https://doi.org/10.1021/acsomega.9b03789.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Zhou JY, Li X, Zheng JY, Dai CC. Volatiles released by endophytic Pseudomonas fluorescens promoting the growth and volatile oil accumulation in Atractylodes lancea. Plant Physiol Biochem. 2016;101:132–40. https://doi.org/10.1016/j.plaphy.2016.01.026.

    Article  CAS  PubMed  Google Scholar 

  172. Yin DD, Wang YL, Yang M, Yin DK, Wang GK, Xu F. Analysis of Chuanxiong Rhizoma substrate on production of ligustrazine in endophytic Bacillus subtilis by ultra high performance liquid chromatography with quadrupole time-of-flight mass spectrometry. J Sep Sci. 2019;42(19):3067–76. https://doi.org/10.1002/jssc.201900030.

    Article  CAS  PubMed  Google Scholar 

  173. Li J, Zhao GZ, Varma A, Qin S, Xiong Z, Huang HY, Zhu WY, Zhao LX, Xu LH, Zhang S, Li WJ. An endophytic Pseudonocardia species induces the production of artemisinin in Artemisia annua. PLoS ONE. 2012;7(12):e51410. https://doi.org/10.1371/journal.pone.0051410.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Saia S, Corrado G, Vitaglione P, Colla G, Bonini P, Giordano M, Stasio ED, Raimondi G, Sacchi R, Rouphael Y. An endophytic fungi-based biostimulant modulates volatile and non-volatile secondary metabolites and yield of greenhouse basil (Ocimum basilicum l.) through variable mechanisms dependent on salinity stress level. Pathogens. 2021;10(7):797. https://doi.org/10.3390/pathogens10070797.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Tiwari R, Awasthi A, Mall M, Shukla AK, Srinivas KS, Syamasundar KV, Kalra A. Bacterial endophyte-mediated enhancement of in planta content of key terpenoid indole alkaloids and growth parameters of Catharanthus roseus. Ind Crops Prod. 2013;43:306–10. https://doi.org/10.1016/j.indcrop.2012.07.045.

    Article  CAS  Google Scholar 

  176. Kusari S, Lamshöft M, Kusari P, Gottfried S, Zühlke S, Louven K, Hentschel U, Kayser O, Spiteller M. Endophytes are hidden producers of maytansine in Putterlickia roots. J Nat Prod. 2014;77(12):2577–84. https://doi.org/10.1021/np500219a.

    Article  CAS  PubMed  Google Scholar 

  177. Kumar A, Singh R, Giri DD, Singh PK, Pandey KD. Effect of Azotobacter chroococcum CL13 inoculation on growth and curcumin content of turmeric (Curcuma longa L.). Int J Curr Microbiol App Sci. 2014;3(9):275–83.

    Google Scholar 

  178. Bonilla A, Sarria ALF, Algar E, Ledesma FM, Solano BR, Fernandes JB, Mañero FG. Microbe associated molecular patterns from rhizosphere bacteria trigger germination and Papaver somniferum metabolism under greenhouse conditions. Plant Physiol Biochem. 2014;74:133–40. https://doi.org/10.1016/j.plaphy.2013.11.012.

    Article  CAS  PubMed  Google Scholar 

  179. Wang Y, Dai CC, Cao JL, Xu DS. Comparison of the effects of fungal endophyte Gilmaniella sp. and its elicitor on Atractylodes lancea plantlets. World J Microbiol Biotechnol. 2012;28:575–84. https://doi.org/10.1007/s11274-011-0850-z.

    Article  PubMed  Google Scholar 

  180. Irmer S, Podzun N, Langel D, Heidemann F, Kaltenegger E, Schemmerling B, Ober D. New aspect of plant–rhizobia interaction: alkaloid biosynthesis in Crotalaria depends on nodulation. Proc Natl Acad Sci. 2015;112(13):4164–9. https://doi.org/10.1073/pnas.1423457112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Scherling C, Ulrich K, Ewald D, Weckwerth W. A metabolic signature of the beneficial interaction of the endophyte Paenibacillus sp. isolate and in vitro–grown poplar plants revealed by metabolomics. Mol Plant Microbe Interact. 2009;22(8):1032–7. https://doi.org/10.1094/MPMI-22-8-1032.

    Article  CAS  PubMed  Google Scholar 

  182. Lòpez-Fernàndez S, Compant S, Vrhovsek U, Bianchedi PL, Sessitsch A, Pertot I, Campisano A. Grapevine colonization by endophytic bacteria shifts secondary metabolism and suggests activation of defense pathways. Plant Soil. 2016;405:155–75. https://doi.org/10.1007/s11104-015-2631-1.

    Article  CAS  Google Scholar 

  183. Jha Y. Endophytic bacteria-mediated regulation of secondary metabolites for the growth induction in Hyptis suaveolens under stress. In: Egamberdieva D, Tiezzi A, editors. Medically important plant biomes source of secondary metabolites. Singapore: Springer Singapore; 2019. p. 277–92. https://doi.org/10.1007/978-981-13-9566-6_12.

    Chapter  Google Scholar 

  184. Mishra A, Singh SP, Mahfooz S, Bhattacharya A, Mishra N, Shirke PA, Nautiyal CS. Bacterial endophytes modulates the withanolide biosynthetic pathway and physiological performance in Withania somnifera under biotic stress. Microbiol Res. 2018;212–213:17–28. https://doi.org/10.1016/j.micres.2018.04.006.

    Article  CAS  PubMed  Google Scholar 

  185. Abdelshafy Mohamad OA, Ma JB, Liu YH, Zhang D, Hua S, Bhute S, Li L. Beneficial endophytic bacterial populations associated with medicinal plant Thymus vulgaris alleviate salt stress and confer resistance to Fusarium oxysporum. Front Plant Sci. 2020;11:47. https://doi.org/10.3389/fpls.2020.00047.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Mastan A, Vivek Babu CS, Hiremath C, Srinivas KVNS, Kumar AN, Kumar JK. Treatments with native Coleus forskohlii endophytes improve fitness and secondary metabolite production of some medicinal and aromatic plants. Int Microbiol. 2020;23(2):345–54. https://doi.org/10.1007/s10123-019-00108-x.

    Article  CAS  PubMed  Google Scholar 

  187. Pullaiah T. Endophytes for the Enhanced growth of Coleus forskohlii and enhanced production of forskolin in forskolin. J Nat Prod. 2022. https://doi.org/10.1007/978-981-19-6521-0_9.

    Article  Google Scholar 

  188. Das A, Kamal S, Shakil NA, Sherameti I, Oelmüller R, Dua M, Varma A. The root endophyte fungus Piriformospora indica leads to early flowering, higher biomass and altered secondary metabolites of the medicinal plant, Coleus forskohlii. Plant Signal Behav. 2012;7(1):103–12. https://doi.org/10.4161/psb.7.1.18472.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Zhou JY, Sun K, Chen F, Yuan J, Li X, Dai CC. Endophytic Pseudomonas induces metabolic flux changes that enhance medicinal sesquiterpenoid accumulation in Atractylodes lancea. Plant Physiol Biochem. 2018;130:473–81. https://doi.org/10.1016/j.plaphy.2018.07.016.

    Article  CAS  PubMed  Google Scholar 

  190. Kushwaha RK, Singh S, Pandey SS, Rao DV, Nagegowda DA, Kalra A, Vivek Babu CS. Compatibility of inherent fungal endophytes of Withania somnifera with Trichoderma viride and its impact on plant growth and withanolide content. J Plant Growth Regul. 2019;38:1228–42. https://doi.org/10.1007/s00344-019-09928-7.

    Article  CAS  Google Scholar 

  191. Satheesan J, Narayanan AK, Sakunthala M. Induction of root colonization by Piriformospora indica leads to enhanced asiaticoside production in Centella asiatica. Mycorrhiza. 2012;22:195–202. https://doi.org/10.1007/s00572-011-0394-y.

    Article  CAS  PubMed  Google Scholar 

  192. Maggini V, De Leo M, Mengoni A, Gallo ER, Miceli E, Reidel RVB, Biffi S, Pistelli L, Fani R, Firenzuoli F, Bogani P. Plant-endophytes interaction influences the secondary metabolism in Echinacea purpurea (L.) Moench: an in vitro model. Sci Rep. 2017;7(1):16924. https://doi.org/10.1038/s41598-017-17110-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Maggini V, De Leo M, Granchi C, Tuccinardi T, Mengoni A, Gallo ER, Biffi S, Fani R, Pistelli L, Firenzuoli F, Bogani P. The influence of Echinacea purpurea leaf microbiota on chicoric acid level. Sci Rep. 2019;9(1):10897. https://doi.org/10.1038/s41598-019-47329-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Wang Y, Xu L, Gu YQ, Coleman-Derr D. MetaCoMET: a web platform for discovery and visualization of the core microbiome. Bioinform. 2016;32(22):3469–70. https://doi.org/10.1093/bioinformatics/btw507.

    Article  CAS  Google Scholar 

  195. Neu AT, Allen EE, Roy K. Defining and quantifying the core microbiome: challenges and prospects. PNAS. 2021;118(51):e2104429118. https://doi.org/10.1073/pnas.2104429118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Hamonts K, Trivedi P, Garg A, Janitz C, Grinyer J, Holford P, Singh BK. Field study reveals core plant microbiota and relative importance of their drivers. Environ Microbiol. 2018;20(1):124–40. https://doi.org/10.1111/1462-2920.14031.

    Article  CAS  PubMed  Google Scholar 

  197. Barnett SE, Cala AR, Hansen JL, Crawford J, Viands DR, Smart LB, Smart CD, Buckley DH. Evaluating the microbiome of hemp. Phytobiomes J. 2020. https://doi.org/10.1094/PBIOMES-06-20-0046-R.

    Article  Google Scholar 

  198. Qiu Z, Verma JP, Liu H, Wang J, Batista BD, Kaur S, Singh BK. Response of the plant core microbiome to Fusarium oxysporum infection and identification of the pathobiome. Environ Microbiol. 2022;24(10):4652–69. https://doi.org/10.1111/1462-2920.16194.

    Article  CAS  PubMed  Google Scholar 

  199. Shade A, Handelsman J. Beyond the Venn diagram: the hunt for a core microbiome. Environ Microbiol. 2012;14(1):4–12. https://doi.org/10.1111/j.1462-2920.2011.02585.x.

    Article  CAS  PubMed  Google Scholar 

  200. Berg G, Rybakova D, Fischer D, Cernava T, Vergès MCC, Charles T, Schloter M. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8:1–22. https://doi.org/10.1186/s40168-020-00875-0.

    Article  Google Scholar 

  201. Müller H, Berg C, Landa BB, Auerbach A, Moissl-Eichinger C, Berg G. Plant genotype-specific archaeal and bacterial endophytes but similar Bacillus antagonists colonize Mediterranean olive trees. Front Microbiol. 2015;6:138. https://doi.org/10.3389/fmicb.2015.00138.

    Article  PubMed  PubMed Central  Google Scholar 

  202. Berg G, Zachow C, Cardinale M, Müller H. Ecology and human pathogenicity of plant-associated bacteria. In: Ehlers R-U, editor. Regulation of biological control agents. Dordrecht: Springer; 2011. p. 175–89.

    Chapter  Google Scholar 

  203. Berg G, Raaijmakers JM. Saving seed microbiomes. ISME J. 2018;12(5):1167–70. https://doi.org/10.1038/s41396-017-0028-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Risely A. Applying the core microbiome to understand host–microbe systems. J Anim Ecol. 2020;89(7):1549–58. https://doi.org/10.1111/1365-2656.13229.

    Article  PubMed  Google Scholar 

  205. Neu AT, Allen EE, Roy K. Defining and quantifying the core microbiome: challenges and prospects. Proc Natl Acad Sci. 2021;118(51):e2104429118. https://doi.org/10.1073/pnas.2104429118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Toju H, Peay KG, Yamamichi M, Narisawa K, Hiruma K, Naito K, Fukuda S, Ushio M, Nakaoka S, Onoda Y, Yoshida K. Core microbiomes for sustainable agroecosystems. Nat Plants. 2018;4(9):733. https://doi.org/10.1038/s41477-018-0139-4.

    Article  PubMed  Google Scholar 

  207. Kumar M, Kumar A, Sahu KP, Patel A, Reddy B, Sheoran N, Charishma K, Rajashekara H, Bhagat S, Rathour R. Deciphering core-microbiome of rice leaf endosphere: revelation by metagenomic and microbiological analysis of aromatic and non-aromatic genotypes grown in three geographical zones. Microbiol Res. 2021;246:126704. https://doi.org/10.1016/j.micres.2021.126704.

    Article  CAS  PubMed  Google Scholar 

  208. Bulgarelli D, Rott M, Schlaeppi K, Loren Ver, van Themaat E, Ahmadinejad N, Assenza F, Rauf P, Huettel B, Reinhardt R, Schmelzer E, Peplies J. Revealing structure and assembly cues for Arabidopsis root-inhabiting bacterial microbiota. Nature. 2012;488(7409):91–5. https://doi.org/10.1038/nature11336.

    Article  CAS  PubMed  Google Scholar 

  209. Lundberg DS, Lebeis SL, Paredes SH, Yourstone S, Gehring J, Malfatti S, Tremblay J, Engelbrektson A, Kunin V, Del Rio TG, Edgar RC, Eickhorst T, Ley RE, Hugenholtz P, Tringe SG, Dangl JL. Defining the core Arabidopsis thaliana root microbiome. Nature. 2012;488(7409):86–90. https://doi.org/10.1038/nature11237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Tian BY, Cao Y, Zhang KQ. Metagenomic insights into communities, functions of endophytes and their associates with infection by root-knot nematode, Meloidogyne incognita, in tomato roots. Sci Rep. 2015;5(1):17087. https://doi.org/10.1038/srep17087.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Tian B, Zhang C, Ye Y, Wen J, Wu Y, Wang H, Li H, Cai S, Cai W, Cheng Z, Lei S. Beneficial traits of bacterial endophytes belonging to the core communities of the tomato root microbiome. Agric Ecosyst Environ. 2017;247:149–56. https://doi.org/10.1016/j.agee.2017.06.041.

    Article  Google Scholar 

  212. Yeoh YK, Paungfoo-Lonhienne C, Dennis PG, Robinson N, Ragan MA, Schmidt S, Hugenholtz P. The core root microbiome of sugarcanes cultivated under varying nitrogen fertilizer application. Environ Microbiol. 2016;18(5):1338–51. https://doi.org/10.1111/1462-2920.12925.

    Article  PubMed  Google Scholar 

  213. de Souza RSC, Okura VK, Armanhi JSL, Jorrín B, Lozano N, Da Silva MJ, Arruda P. Unlocking the bacterial and fungal communities assemblages of sugarcane microbiome. Sci Rep. 2016;6(1):1–15. https://doi.org/10.1038/srep28774.

    Article  CAS  Google Scholar 

  214. Ek-Ramos MJ, Gomez-Flores R, Orozco-Flores AA, Rodríguez-Padilla C, González-Ochoa G, Tamez-Guerra P. Bioactive products from plant-endophytic gram-positive bacteria. Front Microbiol. 2019;10:463. https://doi.org/10.3389/fmicb.2019.00463.

    Article  PubMed  PubMed Central  Google Scholar 

  215. Taghinasab M, Jabaji S. Cannabis microbiome and the role of endophytes in modulating the production of secondary metabolites: an overview. Microorganisms. 2020;8(3):355. https://doi.org/10.3390/microorganisms803035.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Dang H, Zhang T, Wang Z, Li G, Zhao W, Lv X, Zhuang L. Succession of endophytic fungi and arbuscular mycorrhizal fungi associated with the growth of plant and their correlation with secondary metabolites in the roots of plants. BMC Plant Biol. 2021;21:1–6. https://doi.org/10.1186/s12870-021-02942-6.

    Article  CAS  Google Scholar 

  217. Wicaksono WA, Jones EE, Monk J, Ridgway HJ. The bacterial signature of Leptospermum scoparium (Mānuka) reveals core and accessory communities with bioactive properties. PLoS ONE. 2016;11(9):e0163717. https://doi.org/10.1371/journal.pone.0163717.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Maggini V, Bandeira Reidel RV, De Leo M, Mengoni A, Rosaria Gallo E, Miceli E, Biffi S, Fani R, Firenzuoli F, Bogani P, Pistelli L. Volatile profile of Echinacea purpurea plants after in vitro endophyte infection. J Nat Prod. 2020;34(15):2232–7. https://doi.org/10.1080/14786419.2019.1579810.

    Article  CAS  Google Scholar 

  219. Ray S, Singh J, Rajput RS, Yadav S, Singh S, Singh HB. A thorough comprehension of host endophytic interaction entailing the biospherical benefits a metabolomic perspective. In: Jha S, editor. Endophytes and secondary metabolites. Cham: Springer International Publishing; 2019. https://doi.org/10.1007/978-3-319-90484-9_16.

    Chapter  Google Scholar 

  220. Chen H, Wu H, Yan B, Zhao H, Liu F, Zhang H, Liang Z. Core microbiome of medicinal plant Salvia miltiorrhiza seed: a rich reservoir of beneficial microbes for secondary metabolism? Int J Mol Sci. 2018;19(3):672. https://doi.org/10.3390/ijms19030672.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Kumari P, Shanker K, Singh A. Insight into Andrographis paniculata associated bacterial endomicrobiome and assessment of culturable bacterial endophytes for enhancement of industrially important andrographolide content. Ind Crops Prod. 2023;200:116840. https://doi.org/10.1016/j.indcrop.2023.116840.

    Article  CAS  Google Scholar 

  222. Jurburg SD, Eisenhauer N, Buscot F, Chatzinotas A, Chaudhari NM, Heintz-Buschart A, Singh BK. Potential of microbiome-based solutions for agrifood systems. Nat Food. 2022;3(8):557–60. https://doi.org/10.1038/s43016-022-00576-x.

    Article  PubMed  Google Scholar 

  223. Ke J, Wang B, Yoshikuni Y. Microbiome engineering: synthetic biology of plant-associated microbiomes in sustainable agriculture. Trends Biotechnol. 2021;39(3):244–61. https://doi.org/10.1016/j.tibtech.2020.07.008.

    Article  CAS  PubMed  Google Scholar 

  224. Kaul S, Choudhary M, Gupta S, Dhar MK. Engineering host microbiome for crop improvement and sustainable agriculture. Front Microbiol. 2021;12:635917. https://doi.org/10.3389/fmicb.2021.635917.

    Article  PubMed  PubMed Central  Google Scholar 

  225. Mishra AK, Sudalaimuthuasari N, Hazzouri KM, Saeed EE, Shah I, Amiri KM. Tapping into plant–microbiome interactions through the lens of multi-omics techniques. Cell. 2022;11(20):3254. https://doi.org/10.3390/cells11203254.

    Article  CAS  Google Scholar 

  226. Fujiwara F, Miyazawa K, Nihei N, Ichihashi Y. Agroecosystem engineering extended from plant-microbe interactions revealed by multi-omics data. Biosci Biotechnol Biochem. 2023;87(1):21–7. https://doi.org/10.1093/bbb/zbac191.

    Article  Google Scholar 

  227. Lawson CE, Harcombe WR, Hatzenpichler R, Lindemann SR, Löffler FE, O’Malley MA, García Martín H, Pfleger BF, Raskin L, Venturelli OS, Weissbrodt DG, Noguera DR, McMahon KD. Common principles and best practices for engineering microbiomes. Nat Rev Microbiol. 2019;17(12):725–41. https://doi.org/10.1038/s41579-019-0255-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Gopalakrishnappa C, Gowda K, Prabhakara KH, Kuehn S. An ensemble approach to the structure-function problem in microbial communities. Iscience. 2022;25(2):103761.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. San León D, Nogales J. Toward merging bottom–up and top–down model-based designing of synthetic microbial communities. Curr Opin Microbiol. 2022;69:102169. https://doi.org/10.1016/j.mib.2022.102169.

    Article  PubMed  Google Scholar 

  230. Verstraete W, Wittebolle L, Heylen K, Vanparys B, De Vos P, Van de Wiele T, Boon N. Microbial resource management: the road to go for environmental biotechnology. Eng Life Sci. 2007;7(2):117–26. https://doi.org/10.1002/elsc.200620176.

    Article  CAS  Google Scholar 

  231. Albertsen M, Hugenholtz P, Skarshewski A, Nielsen KL, Tyson GW, Nielsen PH. Genome sequences of rare, uncultured bacteria obtained by differential coverage binning of multiple metagenomes. Nat Biotechnol. 2013;31:533–8. https://doi.org/10.1038/nbt.2579.

    Article  CAS  PubMed  Google Scholar 

  232. Wang L, Dash S, Ng CY, Maranas CD. A review of computational tools for design and reconstruction of metabolic pathways. Synth Syst Biotechnol. 2017;2:243–52. https://doi.org/10.1016/j.synbio.2017.11.002.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Zomorrodi AR, Segre D. Synthetic ecology of microbes: mathematical models and applications. J Mol Biol. 2015;428:837–86. https://doi.org/10.1016/j.jmb.2015.10.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Borenstein E, Kupiec M, Feldman MW, Ruppin E. Large-scale reconstruction and phylogenetic analysis of metabolic environments. Proc Natl Acad Sci. 2008;105(38):14482–7. https://doi.org/10.1073/pnas.0806162105.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Jiang CY, Dong L, Zhao JK, Hu X, Shen C, Qiao Y, Zhang X, Wang Y, Ismagilov RF, Liu SJ, Du W. High-throughput single-cell cultivation on microfluidic streak plates. Appl Environ Microbiol. 2016;82(7):2210–8. https://doi.org/10.1128/AEM.03588-15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Browne HP, Forster SC, Anonye BO, Kumar N, Neville BA, Stares MD, Goulding D, Lawley TD. Culturing of ‘unculturable’ human microbiota reveals novel taxa and extensive sporulation. Nature. 2016;533(7604):543–6. https://doi.org/10.1038/nature17645.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Xu P, Clark C, Ryder T, Sparks C, Zhou J, Wang M, Russell R, Scott C. Characterization of TAP Ambr 250 disposable bioreactors, as a reliable scale-down model for biologics process development. Biotechnol Prog. 2017;33(2):478–89. https://doi.org/10.1002/btpr.2417.

    Article  CAS  PubMed  Google Scholar 

  238. Song Y, Yin H, Huang WE. Raman activated cell sorting. Curr Opin Chem Biol. 2016;33:1–8. https://doi.org/10.1016/j.cbpa.2016.04.002.

    Article  CAS  PubMed  Google Scholar 

  239. Hatzenpichler R, Connon SA, Goudeau D, Malmstrom RR, Woyke T, Orphan VJ. Visualizing in situ translational activity for identifying and sorting slow-growing archaeal−bacterial consortia. Proc Natl Acad Sci. 2016;113(28):E4069–78. https://doi.org/10.1073/pnas.1603757113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Karimi A, Karig D, Kumar A, Ardekani AM. Interplay of physical mechanisms and biofilm processes: review of microfluidic methods. Lab Chip. 2015;15:23–42. https://doi.org/10.1039/C4LC01095G.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Aleklett K, Kiers ET, Ohlsson P, Shimizu TS, Caldas VE, Hammer EC. Build your own soil: exploring microfluidics to create microbial habitat structures. Isme J. 2018;12:312–9. https://doi.org/10.1038/ismej.2017.184.

    Article  PubMed  Google Scholar 

  242. He Z, Wu H, Yan X, Liu W. Recent advances in droplet microfluidics for microbiology. Chin Chem Lett. 2022;33(4):1729–42. https://doi.org/10.1016/j.cclet.2021.08.059.

    Article  CAS  Google Scholar 

  243. Gach PC, Shih SC, Sustarich J, Keasling JD, Hillson NJ, Adams PD, Singh AK. A droplet microfluidic platform for automating genetic engineering. ACS Synth Biol. 2016;5(5):426–33. https://doi.org/10.1021/acssynbio.6b00011.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

PK and ND acknowledge the University Grants Commission (UGC) Senior Research Fellowship. VS acknowledges support from the European Commission (Horizon 2020 FET-OPEN Grant Agreement No. 828940) and the Swedish Research Council FORMAS (Grant #2019-00912). The authors would also like to acknowledge Biorender.com for designing the figure templates. The publication number provided by the institute is CIMAP/PUB/2023/160.

Funding

Open access funding provided by Royal Institute of Technology. This work was supported by funding to A.S. from the Council of Scientific and Industrial Research (CSIR) through (Plg/MLP0048/NCP-FBR 2020-CSIR-NBRI) and Science and Engineering Board (SERB), Department of Science and Technology, Government of India (Grant Number: SPG/2021/003168).

Author information

Authors and Affiliations

Authors

Contributions

AS conceived the initial idea. PK, ND, PKT, BKS, VS, and AS wrote and reviewed the manuscript. PK and ND prepared figures and tables. All authors read and approved the final version of the manuscript.

Corresponding authors

Correspondence to Vaibhav Srivastava or Akanksha Singh.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The original online version of this article was revised: The affiliation details for the corresponding author, Vaibhav Srivastava were incorrectly given as '5, 6' but should have been '5'.

Supplementary Information

Additional file 1: Table S1.

List of beneficial endophytic microbes associated with various medicinal plants and their plant growth promoting (PGP) and antimicrobial properties. Table S2. List of endophytes enhancing pharmaceutically important secondary metabolites in medicinal plants.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kumari, P., Deepa, N., Trivedi, P.K. et al. Plants and endophytes interaction: a “secret wedlock” for sustainable biosynthesis of pharmaceutically important secondary metabolites. Microb Cell Fact 22, 226 (2023). https://doi.org/10.1186/s12934-023-02234-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12934-023-02234-8

Keywords