Skip to main content
3 Biotech logoLink to 3 Biotech
. 2023 May 14;13(6):191. doi: 10.1007/s13205-023-03605-z

Endophytic fungi mediates production of bioactive secondary metabolites via modulation of genes involved in key metabolic pathways and their contribution in different biotechnological sector

Prabha Toppo 1, Lahasang Lamu Kagatay 1, Ankita Gurung 1, Priyanka Singla 2, Rakhi Chakraborty 3, Swarnendu Roy 4, Piyush Mathur 1,
PMCID: PMC10183385  PMID: 37197561

Abstract

Endophytic fungi stimulate the production of an enormous number of bioactive metabolites in medicinal plants and affect the different steps of biosynthetic pathways of these secondary metabolites. Endophytic fungi possess a number of biosynthetic gene clusters that possess genes for various enzymes, transcription factors, etc., in their genome responsible for the production of secondary metabolites. Additionally, endophytic fungi also modulate the expression of various genes responsible for the synthesis of key enzymes involved in metabolic pathways of such as HMGR, DXR, etc. involved in the production of a large number of phenolic compounds as well as regulate the expression of genes involved in the production of alkaloids and terpenoids in different plants. This review aims to provide a comprehensive overview of gene expression related to endophytes and their impact on metabolic pathways. Additionally, this review will emphasize the studies done to isolate these secondary metabolites from endophytic fungi in large quantities and assess their bioactivity. Due to ease in synthesis of secondary metabolites and their huge application in the medical industry, these bioactive metabolites are now being extracted from strains of these endophytic fungi commercially. Apart from their application in the pharmaceutical industry, most of these metabolites extracted from endophytic fungi also possess plant growth-promoting ability, bioremediation potential, novel bio control agents, sources of anti-oxidants, etc. The review will comprehensively shed a light on the biotechnological application of these fungal metabolites at the industrial level.

Keywords: Anti-microbial, Anti-oxidants, Bioremediation, Biostimulants, Pharmaceutical

Introduction

Endophytic fungi are among the most diverse group of endophytes that colonize a large number of medicinal plants and have been shown to alter secondary metabolite production (Das et al. 2012; Bajaj et al. 2018). Most of these secondary metabolites are responsible for imparting pharmaceutical properties to medicinal plants (Gupta and Chaturvedi 2019; Hussain et al. 2021a, b). Extensive literature is now available that depicts the role of endophytic fungi associated with medicinal plants in the augmentation of these secondary metabolites (Erb and Kliebenstein 2020; Kong et al. 2021; Yang et al. 2020a, b; Ravi et al. 2022). Endophytic fungi affect the different steps of biosynthetic pathways of these secondary metabolites by modulation of various genes responsible for the synthesis of key enzymes involved in these pathways. Some reports have shown that endophytic fungi modulate the expression of genes HMGR (3-hydroxy-3-methyl-glutaryl-CoA reductase), DXR ((1-deoxy-d-xylulose 5-phosphate reductoisomerase), etc., involved in the production of a large number of phenolic compounds (Ming et al. 2013; Zhai et al. 2018). Furthermore, endophytic fungi also regulate the expression of genes involved in the production of alkaloids and terpenoids (Pandey et al. 2016a, b; Singh et al. 2020a). The present review will give detailed information regarding the endophyte-mediated gene expression of these metabolic pathways.

Endophytic fungi stimulate the production of an enormous number of bioactive metabolites as evident from studies carried out in previous years (Nischitha and Shivanna 2022; Han et al. 2022; Mohamed et al. 2022; Siebatcheu et al. 2022; Kalimuthu et al. 2022). Studies have been carried out for the isolation of these secondary metabolites from these endophytic fungi at a large scale and for analyzing their bioactivity (Zhu et al. 2022; Siebatcheu et al. 2022). Substantial reports of bioactivity of these metabolites have been observed such as anti-microbial, anti-cancerous, anti-inflammatory, anti-viral, anti-oxidative, etc. (Jayasekara et al. 2022; Bedi et al. 2018; Maharjan et al. 2020). Due to the ease of synthesis and their huge application in the medical industry, a number of bioactive metabolites such as Fumitremorgin D, Penochalasin I, 3-O-methylfunicone, Herquline B, Ferrirubin, Peniproline A, Saponins (Liang et al. 2015; Huang et al. 2016; Wang et al. 2017; Jin et al. 2017) are now being extracted from various endophytic fungi such as Aspergillus, Trichoderma, Penicillium, Fusarium, Phomopsis etc. and are are being commercially utilized for their applications in the biomedicine, agriculture, food, and biotechnology industries (El-Sayed et al. 2020; Torres-Mendoza et al. 2020; Devi et al. 2023a).

Fungal endophytes have emerged as a key player in the biotechnological sector apart from their application in the pharmaceutical industry, most of these endophytes have been shown to bear plant growth-promoting ability, bioremediation potential, as well as novel biocontrol agents (Wang et al. 2021; d’Errico et al. 2021). The present review will elaborate on all these aspects of endophytic fungi and discuss their modern-day application in the biotechnology sector.

Most of the previous articles published concerning endophytic fungi have not shown the effect of the endophytic fungi on the host at the molecular level that leads to secondary metabolite production. This review will be the first of its kind that comprehensively discusses the endophytic-mediated secondary metabolites and their molecular mechanism for this increased concentration of secondary metabolites.

Plant secondary metabolites

Plant secondary metabolites have been classified according to their chemical structure into several major classes viz.-terpenoids, phenylpropanoids (phenolic substances), nitrogen-containing compounds (alkaloids, non-protein amino acids, and cyanogenic glycosides), sulfur-containing compounds (phytoalexins, glucosinolates, and defensin) and phytohormones (Erb and Kliebenstein 2020). Phenolics impart significant contributions to pharmacological activities (anti-microbial, anti-inflammatory, anti-diabetic, insecticidal, anti-septic, anti-oxidant, hepatoprotective, anti-coagulant, anti-cancer, etc.) of several medicinal plants (Hussein and El-Anssary 2019). Alkaloids are nitrogen-containing organic compounds that serve as nitrogen reservoirs during environmental stress and actively participate in stress signaling (Bhambhani et al. 2021). Many alkaloids also showed promising anti-microbial (protoberberine from Berberis aristate, tomatidine from tomato piperine from Piper), insecticidal (nicotine from Nicotiana, colchicine from Colchicum autumnale), antiherbivoral (solasonine and solamargine from Solanum tuberosum, swainsonine from Astragalus and Oxytropis), anticarcinogenic (berberine, evodiamine from Evodia rutaecarpa), neuroprotective (rescinnamine from Rauvolfia reflexa, harmaline from Peganum harmala) and other pharmacological properties (Wang et al. 2010; Cretton et al. 2016; Yang et al. 2015; Kong et al. 2021).

Terpenoids or isoprenoids contribute to aroma, fragrance, flavor, and pigments and exert a wide range of pharmacological activities- anti-tumor and anti-cancer (geraniol from Geranium, ursolic bioactive acid from Rosmarinus, cucurbitacin from Cucurbita), anti-inflammatory (paeoniflorin from Paeonia lactiflora, Triptolidenol from Tripterygium wilfordii), anti-microbial (menthol from Mentha, oleanolic acid from Syzygium), antiviral (andrographolide from Andrographis paniculata, betulinic acid from Syzygium claviforum), cardioprotective (Tanshinone from Salvia miltiorrhiza, ginsenoside from Panax ginseng), anti-diabetic (stevioside from Stevia rebaudiana, artimisinin from Artemisia annua), etc. (Yang et al. 2020a, b; Galle et al. 2014; Das 2015; Kim et al. 2017). Non-protein amino acids such as γ-aminobutyric acid (GABA), β-aminobutyric acid (BABA), l-mimosine (from Mimosa pudica), l-DOPA, l-canavanine (from Canavalia ensiformis), etc., showed significant anti-microbial and anti-oxidant activities (Rodrigues-Correa and Fett-Nato 2019). Glucosinolates are abundant in cruciferous plants and play important role in regulating oxidative stress, inhibiting angiogenesis and metastasis, and preventing inflammation along with anti-cancer and anti-bacterial properties (Traka 2016). Venieraki et al. (2017) has reviewed upon the list of well known medicinal plants that are found associated with endophytic fungi and have been employed for the extraction of bioactive secondary metabolites similar to that of medicinal plants.

Endophytic fungi and medicinal plants

Endophytes that are found associated with medicinal plants have been proven to be a source of a large number of bioactive compounds, enzymes, biostimulants, etc. Additionally, these endophytes carry out a versatile role in the production of secondary metabolites in medicinal plants as evidenced by the reports of previous studies (Fig. 1). Several fungal endophytes were isolated from the leaf and stem of a medicinal herb (Ocimum sanctum) collected from three different geographical locations in India and Worldwide. Among the different isolates, Macrophomina phaseolina showed the highest anti-fungal activity against phytopathogenic fungi Sclerotinia sclerotiorum, Rhizoctonia solani, and Fusarium oxysporum (Chowdhary and Kaushik 2015). In a similar type of study, endophytes that have been isolated from four medicinal plants Rheum emodi, Hypericum perforatum, Dioscorea deltoidea, and Artemisia annua in the Jammu and Kashmir region, India showed significant anti-fungal and anti-bacterial activity (Dar et al. 2017). Substantial anti-microbial activity was also observed from the endophytes isolated from the leaves and bark of the traditional endemic medicinal plant Litsea cubeba in the Northeast region of India. The isolates were identified as Nigrospora sphaerica, Acremonium falciforme, Penicillium, chrysogenum, and Allomyces arbuscula. However, A. falciforme was the most prevailing and demonstrate the highest anti-microbial (Deka and Jha 2018). Several fungal endophytes isolated from leaves, flowers, and fruits of Monarda citriodora, an important medicinal and aromatic herb revealed biocontrol potential against many phytopathogenic fungi (Katoch and Pull 2017). Phytopathogenic strains of endophytic fungi viz. Colletotrichum, Eupenicillium, Fusarium, Hypoxylon, Penicillium, Phomopsis, Trametes, Trichoderma, Umbelopsis, Verticillium, and Xylaria that possess anti-microbial activity were isolated from medicinal plant Kadsura angustifolia (Huang et al. 2015).

Fig. 1.

Fig. 1

Schematic representation of a procedure for isolation and identification of the endophytic fungi from different parts of medicinal plants. Endophytic fungi act as bioinoculant for augmenting the growth and development of plants and modulate the transcript expression of genes involved in the biosynthetic pathway of secondary metabolite production. (ANR- Anthocyanidin reductase; HDR- 1-hydroxy-2-methyl-2-(E)-butenyl-4-diphosphate reductase; HMGR- 3-Hydroxy-3-Methylglutaryl-CoA Reductase; HMGS- 3-Hydroxy-3-methylglutaryl-CoA synthase; 4CL- 4-coumarate:Co1-ligase; C4H- Cinnamate-4-hydroxylase; CHI- Chalcone isomerase; CHS-Chalcone synthase; CMK- 4-diphosphocytidyl-2-C-methyl-D-erythritol kinase; DFR- Dihydroflavonol 4-reductase; DXR- 1-deoxy-D-xylulose 5-phosphate reductor isomerase; DXS- 1-deoxy-D-xylulose-5-phosphate synthase; F3H:Flavonoid 3-hydroxylase;H6H- Hyoscyamine 6β-hydroxylase PAL- Phenylalanine ammonia lyase; PMK- Phosphomevalonate kinase; ODC- Ornithine decarboxylases; PMT- Putrescine N-methyltransferase; QPT- Quinolinic acid phosphoribosyl Transferase; SMT1- C-24 sterol methyltransferase 1; SMT2- C-24 sterol methyltransferase 2; TR 1- Tropinone reductase 1)

Besides that, it has been reported that endophytic fungi (Alternaria alternata and Fusarium sp.) isolated from the medicinal plant Mappia foetida possess potent anti-cancerous activity (Ravi et al. 2022). GC–MS analysis confirmed the presence of two important anti-cancerous compounds triruthenium dodecacarbonyl and molybdenum in fungal extracts synthesized from the above endophytic fungi (Ravi et al. 2022). Similarly, endophytic fungi isolated from different medicinal plants in their extract showed the presence of different phenolic substances like catechol, p-OH benzoic, ferulic, protocatechuic acids, and cinnamic acid (Hassane et al. 2022). Additionally, different flavonoids were also detected such as chrysin, rutin, apigenin, luteolin, acacetin, luteolin, and quercetin (Hassane et al. 2022). Similarly, endophytic fungi Nigrospora sphaerica isolated from the medicinal plant Euphorbia hirta (dudhi) L. revealed the occurrence of 2,4-Di-tert-butylphenol in the ethyl acetate fungal extract which is a phenolic compound liable for the anti-oxidative potential (Gautam et al. 2022). Concomitantly, the endophytes under culture showed the presence of tannins, phenols, ascorbic acid, flavonoids, etc. which indicated their potential for extraction of bioactive compounds (Kumaresan et al. 2015). Several different endophytic fungi were isolated from different parts of the medicinal tree Aegle marmelos grown in the Western Ghats of India. Some endophytic fungi were pigmented and were identified as Cladosporium cladosporioides FC75A, Aspergillus niger FC30AGr, Alternaria alternate FC39BY, Curvularia australiensis FC2AP, Alternaria citrimacularis FC8ABr. Bioactive metabolites extracted from this fungus showed noteworthy anti-oxidant and anti-microbial activity against many bacterial and fungal pathogens under in-vitro assays (Mani et al. 2015). Another study by Sheik and Chandrashekar (2018) also reported anti-microbial and free radical scavenging activity from the endophytic fungi isolated from the endemic plant of Humboldtia brunonis. The study also showed variation in endophyte frequency over different seasons. An endophytic fungus Curvularia geniculata isolated from the medicinal plant Phyllanthus niruri was assessed for anti-oxidant potential and anti-cancer activity (Kalimuthu et al. 2022). In another work, the endophytic fungus Aspergillus fumigatus isolated from traditional Chinese medicine, Crocus sativus showed the presence of thirty-two different chemical compounds that were reported to be secondary metabolites namely, anthraquinones, emodin, physcion, carviolin, endocrocin, alkaloids, pseurotin A, fumiquinazoline C, two benzoate derivatives, methyl asterrate, and asterric acid (Jiang et al. 2022).

Studies have also shown that the endophytic fungi isolated from medicinal plants improve the growth of the host plants by producing ammonia, solubilizing phosphates, siderophore production as well as indole acetic acid (IAA) production (Toppo et al. 2022; Huang et al. 2022; Zhu et al. 2022; Madasi et al. 2021). Chen et al. (2021a) reported in their study that endophytic fungi Byssochlamys spectabilis 1-N2, Chaetomium nigricolor 3-G7, and Phomopsis sp.1-G1 isolated from medicinal plant Bletilla striata were capable of producing IAA, siderophore and cellulolytic enzymes that significantly increased the growth of the host plants. Furthermore, from the leaves of medicinal plants Ephedra pachyclada, fifteen fungal endophytes belonging to the genera Penicillium, Alternaria, and Aspergillus sp. were isolated and tested for their plant growth promotion activity, enzyme production as well as anti-microbial activity (Khalil et al. 2021). In a different study, Khan et al. (2021) isolated the fungal endophyte Acremonium sp. Ld-03 from the bulbs of the Chinese medicinal plant Lilium davidii. The endophytic fungal strain Ld-03 showed significant production of IAA, siderophore, and phosphate solubilization activity. Further, in vitro analysis of the growth of Allium tuberosum was assessed in presence of endophytic fungal strain Ld-03 and it was observed that plants tend to increase in root and shoot length. A study showed that endophytic fungi (Colletotrichum sp. SXS649, Pestalotiopsis sp. SXS650, Diaporthe sp. SXS652, and Botryosphaeriales SXS651) isolated from a native medicinal plant of the Brazilian savanna, i.e., Palicourea rigida exhibited substantial production of amylase, cellulase, protease, and tannase (Dos Santos et al. 2021). Moreover, the thirty-two fungal endophytes isolated from the stem and bark of Albizia lebbeck showed a positive result for the extracellular production of all enzymes such as amylase, protease, lipase, laccase, cellulase, and gelatinase (Mathur et al. 2022). Additionally, the utilization of these enzymes in the biodegradation of vegetable oil, dye decolourization, saccharification of bagasse and paper, and wastewater treatment was also observed (Mathur et al. 2022). A large endophytic fungal diversity has been isolated from the leaves of the medicinal plants of Brazil Myracrodruon urundeuva. Among the different isolates, the isolate Diaporthe was unique in the sense that it produces the important amino acid l-asparagine (Pádua et al. 2019). Several studies showing the number of endophytic fungi isolated from different medicinal plants have been summarized in Table 1.

Table 1.

A list of endophytic fungi with potential bioactivity isolated from different parts of medicinal plants

Medicinal plant Host tissue Endophytic fungi Bioactivity References
Bauhinia forficata

Leaves

Sepals

Stems

Seeds

Acremonium curvulum

Aspergillus ochraceus

Gibberella fujikuroi

Myrothecium verrucaria

Trichoderma piluliferum

Anti-biotic

Enzyme source

Bezerra et al. (2015)
Ocimum sanctum

Leaves

Stems

Fusarium verticillioides

Hypocrea sp.

Bipolaris maydis

Macrophomina phaseolina

Meyerozyma guilliermondii

Chaetomium coarctatum

Rhizoctonia bataticola Hypoxylon sp.

Diaporthe phaseolorum

Alternaria tenuissima

A. alternate

Anti-phytopathogenic Chowdhary and Kaushik (2015)
Asclepias sinaica Leaves

Penicillium chrysogenum

Alternaria alternata

Anti-microbial

Enzyme source

Fouda et al. (2015)

Calotropis procera, Catharanthus roseus

Euphorbia prostrate Vernonia amygdalina Trigonella foenum-graecum

Leaves

Stems

Seeds

Alternaria sp.

Bipolaris sp.

Curvularia sp.

Chaetomium sp.

Drechslera sp.

Emericella sp.

Aspergillus sp.

Cladosporium sp.

Paecilomyces sp.

Phoma sp.

Anti-oxidant Khiralla et al. (2015)
Azadirachta indica Leaves

Chaetomium sp.

Curvularia sp.

Colletotrichum sp.

Trichoderma sp.

Kumaresan et al. (2015)
Aegle marmelos

Barks,

Branches

Leaves

Roots

Alternaria alternata,

Alternaria citrimacularis

Curvularia australiensis

Anti-microbial Mani et al. (2015)
Rauwolfia serpentine

Leaves

Stems

Colletotrichum gloeosporioides

Penicillium sp.

Aspergillus sp.

Anti-microbial Anti-oxidant

Hypocholestraemic

Nath et al. (2015)
Solanum xanthocarpum Leaves Phomopsis vexans Drug-Lovastin Parthasarathy and Sathiyabama (2015)
Crescentia cujete Leaves

Nigrospora sphaerica

Fusarium oxysporum Gibberella moniliformis

Beauveria bassiana

Anti-bacterial Anti-cancerous

Anti-oxidant

Prabukumar et al. (2015)

Ocimum sanctum

Aloe vera

Leaves

Stems

Roots

18 endophytic fungi were isolated

Anti-fungal

Enzyme source

Yadav et al. (2016)

Anthocleista djalonensis

Fagara zanthoxyloides

Leaves

Colletotrichum gloeosporioides

Pestalotiopsis thea

Anti-Respiratory Syncytial Virus Compounds Uzor et al. (2016)

Calotropis procera Trigonella foenum-graecum

Vernonia amygdalina

Catharanthus roseus Euphorbia prostrata

Leaves

Stems

Seeds

Alternaria alternata

Aspergillus terreus Cladosporium cladosporioides

Trametes versicolor Chaetomium globosum Aspergillus terreus

Hansfordia sinuosae

Curvularia aeria

Pleosporales sp.

Phoma multirostrata

Curvularia australiensis

Alternaria sp.

Byssochlamys spectabilis

Cytotoxic

Antibiotic

Khiralla et al. (2016)

Nothapodytes foetida

Hypericum mysorense H. japonicum

Leaves

Stems

Penicillium sp.

Aspergillus sp.

Fusarium sp.

Gliocladium sp.

Cladosporium sp.

Trichoderma sp.

Colletotrichum sp.

Pestalotiopsis sp.

Anti-oxidant

Free radical scavenging

Samaga and Rai (2016)
Combretum lanceolatum Roots

Trichoderma spirale

Penicillium verruculosum

Penicillium simplicissimum

Fusarium oxysporum

Diaporthe phaseolorum

Leptosphaerulina chartarum Cladosporium perangustum

Anti-microbial

Anti-oxidant

de Siqueira et al. (2017)
Monarda citriodora

Leaves

Roots

Flowers

Fusarium sp.

Aspergillus sp.

Cladosporium sp.

Biocontrol Katoch and Pull (2017)
Mirabilis jalapa Roots Aspergillus clavatonanicu Anti-microbial Mishra et al. (2017)
Elaeocarpus sylvestris

Leaves

Stems

Pestalotiopsis sp.

Diaporthales sp.

Meyerozyma sp.

Diaporthales sp.

Pestalotiopsis sp.

Pseudocercospora sp.

Anti-oxidants Prihantini and Tachibana (2017)
Glycyrrhiza glabra Rhizomes Diaporthe terebinthifolii

Diapolic acid A–B

Cytotoxic Anti-microbial activity

Yedukondalu et al. (2017)
Humboldtia brunonis

Leaves

Stems

Aspergillus sp.

Curvularia clavata

Curvularia pallescens

Debaromyces hansenii Guignardia sp.

Hypoxylon anthochroum Meyerozyma caribbica Paecilomyces lilacinus Alternaria alternate

Cunninghamella echinulata Fusarium fusarioides

Fusarium oxysporum

Phanerochaete sp.

Anti-microbial

Free radical scavenging

Sheik and Chandrashekar (2018)
18 medicinal plants

Leaves

Stems

Aspergillus flavus

A. niger

A. sydowii

A. versicolor

Aspergillus sp.

Anti-viral

Anti-oxidant

Selim et al. (2018)
Terminalia pallida Rhynchosia beddomei Pterocarpus santalinus

Leaves

Stems

Cladosporium delicatulum Plumbagin-Anti-microbial Venkateswarulu et al. (2018)
Hypericum perforatum

Leaves

Stems

Roots

Flowers

Epicoccum nigrum

Alternaria sp.

Trichoderma harzianum

Anti-microbial Vigneshwari et al. (2019)

Zingiber officinale

Salix sp.

Leaves Epicoccum nigrum

Anti-fungal

Anti-bacterial

Cytotoxic potential

Harwoko et al. (2019)
Aloe dhufarensis Lavranos Leaves Sarocladium kiliense Penicillium oxalicum Anti-phytopathogenic Al-Rashdi et al. (2020)

Cotyledon orbiculata

Psychotria zombamontana,

Tecomaria capensis

Catha edulis

Melianthus comosus

Leaves

Stems

Roots

Diaporthe sp.

Talaromyces funiculosus

Penicillium commune Cochliobolus sp.

Phomopsis sp.

Clonostachys rosea

Anti-microbial

Enzyme source

Presence of type I polyketide synthases (PKS)

Abdalla et al. (2020)

Papaver somniferum

Cassia fistula

Catharanthus roseus

Roots

Leaves

Aspergillus sp.

Curvularia sp.

Anti-bacterial

Anti-fungal

Khattak et al. (2020)
Aquilaria malaccensis

Stem

Leaves

Alternaria sp.

Curvularia sp.

Rhizopus sp.

Sterilia sp.

Anti-microbial Mochahari et al. (2020)

Lafoensia pacari

Guazuma ulmifolia Campomanesia xanthocarpa

Siparuna guianensis

Leaves

Colletotrichum sp.

Diaporthe sp.

Bjerkandera sp.

Talaromyces sp.

Cochliobolus sp.

Phaeophlebiopsis sp.

Curvularia sp.

Anti-oxidants

Anti-inflammatory

Santos et al. (2020)
Memecylon umbellatum

Leaves

Bark

Fruits

Endomelanconiopsis endophytica

Lasiodiplodia iranensis

L. theobromae

Phyllosticta capitalensis Aspergillus niger

Cochlibolus lunatus Pestalotiopsis guepinii

Anti-bacterial Suryavamshi and Shivanna (2020)
Euphorbia geniculate

Leaves

Stems

Roots

Aspergillus favus

A. ochraceus

A. terreus

Emercilla nidulans var. acristata

Macrophomina phaseolina

Anti-fungal Kamel et al. (2020)
Taxus baccata Bark

Acremonium Colletotrichum Fusarium sp.

Nodulisporium sp.

Paecilomyces

Periconia sp.

Paclitaxel

Cytotoxic

El-Bialy and El-Bastawisy (2020)
Euphorbia larica Roots

Neocosmospora sp.

Alternaria alternata

Anti-fungal Khuseib et al. (2020)
Siraitia grosvenorii

Roots

Stems

Leaves

Fruits

Diaporthe angelica

Fusarium solani

Mogroside V Bin et al. (2020)

Endophytic fungi and bioactive metabolite production

Endophytes play a key role in the production of a large number of bioactive metabolites and bioprospecting of endophytes in search of novel metabolites has gained considerable attention all over the World (Gouda et al. 2016). Endophytes are the source of new natural compounds including polyketides, alkaloids, phenylpropanoids, terpenoids, and phenols (Fig. 1). Yang et al. (2019) reported the production of a completely new sesquiterpenoid colletotrichine A, a potent inhibitor of acetylcholinesterase (AChE) from endophytic fungus Colletotrichum gloeosporioides GT-7 isolated from the healthy tissues of Uncaria rhynchophylla. The endophytic fungus, Neosartorya fischeri JS0553 is a rich source of new secondary metabolites meroditerpenoid named sartorypyrone E and fischerin compounds and these compounds possess significant neuroprotective activity (Bang et al. 2019). Furthermore, isolated endophytic fungi Eurotium rubrum from the roots and shoots of the mangrove plant Suaeda salsa produces many anthraquinones (such as catenarin, rubrocristin, emodin, and 2-methyleurotinone) that have anti-bacterial, anti-fungal, anti-mycobacterial, anti-malarial and cytotoxic activities (Zhang et al. 2017). Another study reported that an endophytic fungus Mucor irregularis QEN-189, isolated from the fresh inner tissue of the stems of the marine mangrove plant Rhizophora stylosa has potent unique compounds rhizovarins A–F and indole-diterpenes (Gao et al. 2016). These organic compounds hold anti-tumor activity against HL-60 and A-549 cell lines. A fungal endophyte Campylocarpon sp. HDN13-307 associated with the roots of Sonneratia caseolaris exhibited production of a new family of secondary metabolites such as 4-hydroxy-2-pyridone alkaloids, campyridones A and B and campyridones C and D that have strong cytotoxic activity against cancerous cells (Zhu et al. 2016).

An endophytic fungus Neofusicoccum luteum isolated from the plant Kigelia africana led to the formation of a bioactive secondary metabolite C16-terpene dilactole. This metabolite exhibited anti-bacterial activity against Pseudomonas aeruginosa (Bodede et al. 2022). Elkhouly et al. (2021a) reported that the endophytic fungus Aspergillus terreus AH1 isolated from the tissues of Ipomoea carnea produced significant bioactive compounds namely, butyrolactone I, pariplanamide B, pyranterrone D, arenarin A, asterrelenin. These compounds showed anti-microbial activities against several pathogenic microbes such as Staphylococcus aureus and Bacillus subtilis which were gram-positive bacteria as well as the gram-negative bacteria Pseudomonas aeruginosa and Escherichia coli. Among all the compounds produced, butyrolactone and periplanamide B showed anti-biofilm activity to a great extent. Periplanamide B also showed effective activity against cancer cell lines such as HCT116, HepG2 and MCF7. Similarly, another study from Elkhouly et al. (2021b) showed that the endophytic fungus Aspergillus tubenginsis ASH4 which was extracted from the plant Hyoscyamus muticus produced the bioactive metabolite afonic acid. This afonic acid showed acute anti-microbial activity against several bacteria like Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli, and Bacillus subtilis which are very much pathogenic against humans. It also hinders biofilm formation and had anti-oxidant and anti-cancer activities against HCT116, HepG2, and MCF7. GC–MS analysis of fungal extract of endophytic fungi Cladosporium cladosporioides and C. tenuissimum showed the presence of 1-octen-3-ol, 3-methyl-1-butanol, hexanol, trans-2-octanol and hexadecanol. These compounds were cloasely related to the compounds found in host extract, i.e., leaf of Cymbopogon martini and have significant anti-oxidant, anti-cancer and anti-microbial activities (Jayaram et al. 2021). Furthermore, an endophytic fungus Talaromyces assiutensis JTY2 isolated from the leaves of the host plant Ceriops tagal produced secondary metabolite helicascolide F, talaromydine that exhibited both anti-fungal and cytotoxic activities against many phytopathogenic fungi (Li et al. 2021). Another study by Zou et al. (2021) reported that an endophytic fungus Aspergillus fumigatus HQD24 isolated from the flower of Rhizophora mucronata, a Chinese mangrove plant led to the production of 8 alkaloids namely pyripyropene A, 1,11dideacetyl-pyripyropene A, pyripyropene E, chaetominine, tryptoquivaline J, fumitremorgan C, 1-acetyl β-carboline and nicotinic acid. These compounds correspondingly exhibited cytotoxic activity. Among the nine fungal isolates from the pulp of the plant Chaenomeles speciosa, 7 were those of Penicillium spp., one Aspergillus spp., and one Mucor spp. Out of the three fungi, Penicillium spp. produced various bioactive metabolites such as 3-furanacetic acid, 4-hexyl-2,5-dihydro-2,5-dioxo, diisooctyl phthalate, 11-hexadecyn-1-ol and propanedioic acid, dihydroxy that showed anti-microbial activity against Alternaria alternata, Fusarium culmorum and F. oxysporum. Cao et al. (2021) revealed that a natural bioactive product huperzine A produced by the endophytic fungi Penicillium sp. and Colletotrichum gloeosporeoids was found to be a significant remedy against the Alzheimer’s disease. The fungus was isolated from the host plant Huperzia serrata belonging to the family Lycopodiace. Silva et al. (2021) demonstrated the presence of bioactive compounds Mellein and β-orcinaldehyde as the major compound. These were produced from the endophytic fungus Botryossphaeria fabicerciana which was isolated from the leaves of the host plant Morus nigra. These two compounds showed activities against gram-positive bacteria and also possessed anti-oxidative properties. A bis-indole alkaloid, vinblastine was produced by the endophytic fungus Fusarium solani RN1 and Chaetomium funicola RN3 which were isolated from the plant Catharanthus roseus and showed mild anti-cancer activity. The endophytic fungus Alternaria sp. isolated from the plant species Papuacedrus papuana produces tenuazonic acid which is a bioactive metabolite that showed active anti-bacterial activities against Escherichia coli and also showed anti-oxidant activities (Praptiwi et al. 2021). Likewise, Elfita et al. (2022) demonstrated the production of the bioactive compounds 3-hydroxy-4-(hydroxy(4-hydroxyphenyl) methyl) dihyfrofuran-2-on by the fungus Fusarium verticillioides which was isolated from the bark of the host plant Syzygium jambos. The bioactive compound showed anti-bacterial activity against gram-positive bacteria (Staphylococcus aureus, Bacillus subtilis) and gram-negative bacteria (Escherichia coli, Salmonella typhi). The fungal isolate Phyllostica sp. from the medicinal plant Hippobroma longiflora produced useful secondary metabolites such as alkaloids, flavonoids and terpenoids which showed anti-oxidant activities (Widjajanti et al. 2022). A number of studies that summarize the significant role of endophytes in the production of bioactive metabolites have been shown in Table 2.

Table 2.

A list of studies summarizing the fungal endophyte-mediated bioactive secondary metabolite production

Host plant Endophytic fungi Secondary metabolite Bioactivity References
Fritillaria unibracteata var. wabuensis

Fusarium redolens

6WBY3

Peimisine

Imperialine-3b-d-glucoside

Anti-tumor Pan et al. (2015)
Hevea brasiliensis Eutypella scoparia Scoparasin C Cytotoxic Kongprapan et al. (2015)
Diphylleia sinensis Aspergillus fumigatus Fumitremorgin D Cytotoxic Liang et al. (2015)
Myoporum bontioides Penicillium chrysogenum Penochalasin I Cytotoxic Huang et al. (2016)
Arbutus unedo Talaromyces pinophilus

3-O-methylfunicone, Herquline B

Ferrirubin

Insecticidal Vinale et al. (2017b)
Cucumeropsis mannii Penicillium decumbens Peniproline A Cytotoxic Wang et al. (2017)
Panax notoginseng

Fusarium sp.

Aspergillus sp.

Saponins Anti-microbial Jin et al. (2017)
Uncaria rhynchophylla Colletotrichum gloeosporioides Colletotrichine A Acetylcholinesterase inhibitor Chen et al. (2018)
Ficus elastic

Penicillium funiculosum

Trichoderma harzianum

Isocoumarin Anti-microbial Ding et al. (2019)
Vitex negundo Alternaria alternata Alternariol methyl ether (AME) Anti-tumor and anti-cancer Palanichamy et al. (2019)
Glehnia littoralis

Neosartorya

fischeri JS0553

Sartorypyrone E Neuroprotective Bang et al. (2019)
Uvaria grandiflora

Nigrospora spp.

Colletotrichum spp.

Flavonoids

Sterols

Phenols

Terpenoids

Anti-bacterial Anti-oxidant Notarte et al. (2019)
Uncaria rhynchophylla Colletotrichum gloeosporioides

4-Epi-14-hydroxy-10, 23-dihydro-24,25-dehydroaflavinine

10, 23-Dihydro-24,25 -dehydro-21–oxoaflavinine

Ergosterol

Ergosterol peroxide

Mellein

4,5-dihydroblumenol A

Colletotrichine A

Cyclo (L-leucyl-L leucyl)

Brevianamide F

Anti-tumor Yang et al. (2019)
Bruguiera sexangula Phyllosticta capitalensis

Guignardone A

12- hydroxylated guignardone A

Guignardone J

Guignardone M

Xenofuranone B

6,8-dihydroxy-5- methoxy-3-methyl-1H-isochromen-1-one

Regiolone 3,4-dihydroxybenzoic acid

Anti-microbial

Cytotoxic

Xu et al. (2009)
Huperzia serrata Fusarium sp. Rsp5.2 Huperzine A Alzheimer disease Le et al. (2020)
Achyranthes bidentata Phomopsis sp. Chromanones Cytotoxic Yang et al. (2020b)
Phlegmariurus taxifolius Fusarium sp. Huperzine A Alzheimer disease, Acetylcholinesterase inhibition Cruz-Miranda et al. (2020)
Camptotheca acuminate Penicillium polonicum

β-lactone polonicin A

Enoic acid

Polonicin B

Anti-HCC (HepG2 hepatocelluar carcinoma), GLUT4 translocatioGlucose uptake Wen et al. (2020)
Vitex negundo Nigrospora oryzae

Phenol 2,4bis(1,1-dimethylethyl)

3-hexadecyloxycarbonyl-5-(2-hydroxyethyl)-4-methylimidazolium ion

Naphthalene

1,2-benzenedicarboxylic acid diisooctyl ester

Eicosene

Anti-bacterial Suradkar and Hande (2020)
Kigelia africana Neofusicoccum luteum C16-terpene dilactone Anti-bacterial Bodede et al. (2022)
Ipomoea carnea Aspergillus terreus AH1

Butyrolactone I

Pariplanamide B

Pyranterrone D

Arenarin A

Asterrelenin

Anti-microbial, Cytotoxic Anti-biofilm Elkhouly et al. (2021a)
Hyoscyanus muticus Aspergillus tubenginses ASH4 Anofinic acid

Anti-microbial

Anti-biofilm Anti-oxidant Anti-cancer

Elkhouly et al. (2021b)
Cymbopogon martini

Cladosporium clasdosporioides,

Cladosporium tenuissimum

Terpenoid

Alkaloids

Flavonoids

Phenols

Tannins

Anti-oxidant Anti-cancer Anti-microbial Jayaram et al. (2021)
Ceriops tagal Talaromyces assiutensis JTY2

Helicascolide F

Talaromydine

Anti-fungal Cytotoxic Li et al. (2021)
Rhizophora mucronata Aspergillus fumigatus HQD24

Pyripyropene A

1,11dideacetyl-pyripyropene A

Pyripyropene E

Chaetominine

Tryptoquivaline J

Fumitremorgan C

1-acety β-carboline

Nicotinic acid

Cytotoxic Zou et al. (2021)
Chaenomeles speciosa

Penicillium spp.

Aspergillus spp.

Mucor spp.

3-Furanacetic acid

4-hexyl-2,5-dihydro-2,5-dioxo

Diisooctyl phthalate, 11-Hexadecyn-1-ol

Propanedioic acid

dihydroxy

Anti-microbial Lykholat et al. (2021)
Huperzia serrata

Penicillium sp.

Colletotrichum sp.

Gloeosporeoids

Huperzine A Treatment for Alzheimer’s disease Cao et al. (2021)
Morus nigra Botryosphaeria fabicerciana

Mellein,

β-orcinaldehyde

Anti-microbial

Anti-oxidant

Silva et al. (2021)
Catharanthus roseus

Fusarium solani RN1

Chaetomium funicola

Alkaloid

Vinblastine

Vincristine

Anti-cancer Linh et al. (2021)
Lilium davidii Acremonium sp.

Xanthurenic acid

Valyl aspartic acid

Gancidin W

Peptides

Cyclic dipeptides

Valylarginine

Cyclo-[l-(4-hydroxy-Prp)-l-Leu],

Cyclo(pro-phe) (3s,6s)-3-benzyl-6-(4-hydroxybenzyl) piperazine-2,5-dione

Anti-fungal Khan et al. (2021)
Syzygium zeylancium Penicillium brefeldianum p-hydroxybenzaldehyde Anti-bacterial Syarifah et al. (2021)
Papuacedrus papuana Alternaria sp. Tenuazonic acid

Anti-bacterial

Anti-oxidant

Praptiwi et al. (2021)
Eichhornia crassipes Fusarium sp.

Altersolanol

4-hydroxydihydromorjavanicin

5-hydroxy-7-methoxy-2-isopropylchromone

Fusaraichromenone

Anti-microbial Hiranrat et al. (2021)
Acanthus ilicifolius Penicillium sp. Benzophenones Anti-bacterial Bai et al. (2021)
Syzygium jambos Fusarium verticillioides 3-hydroxy-4-(hydroxy(4-hydroxyphenyl) methyl) dihyfrofuran-2-on Anti-bacterial Elfita et al. (2022)
Hippobroma longiflora Phyllosticta sp.

Alkaloid

Flavonoid

Terpenoid

Anti-oxidant Widjajanti et al. (2022)

Endophytic fungi and regulation of key secondary metabolite genes in plants

Ren and Dai (2012) revealed that the endophytic fungus Gilmaniella sp. isolated from the host plant Atractylodes lancea induced the production of jasmonic acid (JA) which acts as a downstream signaling molecule in the NO and H2O2-mediated volatile oil accumulation induced by the endophytic fungi. It was also seen that JA also had interconnection with the salicylic acid (SA) signaling pathway and the expression of the gene HMGR was restored by the JA alongside sesquiterpenoids, which was significantly increased by fungal endophyte. A group of abietanoid diterpenes (tanshinone) were significantly enhanced by the endophytic fungus Trichoderma atroviride D16 found in the host plant Salvia miltiorrhiza due to increased transcript expression of genes HMGR, DXR and GGPPS (geranylgeranyl diphosphate synthase) that plays a major role in the synthesis of tanshinones (Ming et al. 2013) (Fig. 1). Subsequent studies by Chen et al. (2021b), also revealed the up-regulation of the genes HMGR, DXS, DXR, GGPPS, CPS, KSL, and CYP76AHI helped in the biosynthesis of secondary metabolite tanshinone which was produced by the endophytic fungus Cladosporium tenuissimum extracted from the host plant Salvia mitiorrhiza. Similar studies were seen by Zhai et al. (2018), where it was reported that the fungus Chaetomium globosum D38 found in the host plant Salvia mitiorrhiza triggered tanshinones production, a major bioactive compound that was markedly up-regulated by the expression level of the fundamental genes such as HMGR, DXR, GGPPS while CPS and KSL were downregulated. Apart from these, some experimental reports by Verma et al. (2014) showed the biosynthesis of the bioactive secondary metabolites sanguinarin by the increased transcript expression of genes—cheilanthifoline synthase (CFS), stylopine synthase (STS), tetrahydroprotoberberine cis-N-methyltransferase (TNMT), and protopine 6-hydroxylase (P6H). Pandey et al. (2016b) experimentally demonstrated that the fungal endophytes namely Culvularia sp., Choanephora infundibulifera associated with host plant Catharanthus roseus escalated the vindoline content by the upregulation of the genes (geraniol 10-hydroxylase (G10H), tryptophan decarboxylase (TDC), strictosidine synthase (STR), 16-hydoxytabersonine-Omethyltransferase (16OMT), desacetoxyvindoline-4-hydroxylase (D4H), deacetylvindoline-4-Oacetyltransferase (DAT), octadecanoid-responsive Catharanthus AP2-domain protein (ORCA3) (Fig. 1). The gene PRX1 (the gene for vascular class III peroxidase) was also upregulated by the endophyte which is in charge of coupling vindoline and cathranthine and Cys2/His2-type zinc finger protein family transcriptional repressors (ZCTs) were downregulated. Similar reports were made by Magotra et al. (2017) that showed the formation of seven secondary metabolites pseurotin F2, fumagillin, tryprostatin C, pseurotin D, pseurotin A, bis (dimethylthio) gliotoxin, and gliotoxin by the endophytic fungus Aspergillus fumigatus (GA-L7) isolated from the host plant Grewia asiatica of Malvaceae family. There was significant up-regulation in transcript expression of genes Afua_6g 12040, Afua_6g 12050, Afua_6g 12060, Afua_6g 12070, Afua_6g 12080 involved in the biosynthesis of fumiquinazoline C and the presence of valproic acid, an epigenetic modifier upregulated the above genes. Likewise, Yuan et al. (2016) demonstrated increased production of sesquiterpenoids from the ethylene signaling mechanism brought about by the endophytic fungus Gilmaniella sp. AL12 within the host plant Atractylodes lancea via activation of gene TPS21 and TPS11. The endophytic fungus Aspergillus niger CSR3 isolated from the host plant Cannabis sativa modulated the expression of genes P450-1, P450-3, P450-4 which are part of GA biosynthesis pathway and led to increased accumulation of gibberellins (Lubna et al. 2018). Increased transcript of the gene PHGPX by polysaccharide fraction (PSF) induced tanshinones were seen in the case of the endophytic fungus Trichoderma atroviride D16 isolated from the plant Salvia miltiorrhiza belonging to the family Lamiaceae Peng et al. (2019). Several endophytic fungi Culvularia sp., Choamphora infludibulifera, Aspergillus japonicus showed extensive production of root alkaloids ajmalicine and serpentine by modulating the terpenoid indole alkaloid (TIA) pathway in the roots of the plant Catharanthus roseus (Singh et al. 2020a) There was upregulation of the key genes G10H, TDC, and strictosidine synthase and also octadecanoid-derivative responsive Catharanthus AP2/ERF domain transcription activators like ORCA3, while the downregulation of the ZCTs (Cys2/His2-type zinc finger protein family). Ding et al. (2020) reported an active production of a bioactive indole alkaloid meleagrin on the deletion of hdaA (histone deacetylase) an epigenetic regulator in the endophytic fungus isolated from the leaves of Ficus elastic. Therein, there was upregulation of roqO gene which also functioned as the key catalyst in the bioactive secondary metabolite biosynthesis. Table 3 depicts the key genes responsible for the increased production of bioactive metabolites influenced due to endophytic fungi.

Table 3.

A list of studies summarizing the effect of fungal endophyte on bioactive metabolite and regulation of key genes

Name of endophyte Host plant Bioactive metabolites Name of genes Effects of gene References
Gilmaniella sp. Atractylodes lancea Jasmonic acid HMGR Upregulation Ren and Dai. (2012)
Verticillium longisporium Arabidopsis sp. Tryptophan

CYP71A12

CYP71A13

CYP71B15

Upregulation Iven et al. (2012)
Trichoderma atroviride D16 Salvia mitiorrhiza Tanshinone

HMGR

DXR

GGPPS

CPS

KSL

Upregulation-HMGR, DXR, GGPPS

Downregulation- CPS, KSL

Ming et al. (2013)

Chaetomium globosum,

Aspergillus niveoglaucus,

Paecilomyces lilacinus,

Trichoderma harzianum

Papaver somniferum

Sanguinarine,

Shikimate

CFS

STS

TMNT

P6H

Upregulation Verma et al. (2014)

Curvularia sp.,

Choanephora infundibulifera

Catharanthus roseus Vindoline

TDC,

STR,

16OMT,

D4H,

DAT,

ORAC3,

ZTCs,

PRX1

Pandey et al. (2016b)
Aspergillus fumigatus (GA-L7) Grewia asiatica

Pseurotin F2,

Fumagillin,

Tryprostatin C,

Pseurotin D,

Pseurotin A, Bis(dimethylthio)gliotoxin

Gliotoxin

Afua_6g 12040

Afua_6g 12050

Afua_6g 12060

Afua_6g 12070

Afua_6g 12080

Upregulation Magotra et al. (2017)
Gilmaniella sp. Atractylodes lancea Sesquiterpenoids

TPS21,

TPS11

Yuan et al (2016)
Chaetomium globosum D38 Salvia miltiorrhiza

Tanshinones,

Salvianolic acid

HMGR,

DXR,

GGPPS,

CPS,

KSL

Upregulation-HMGR, DXR

Downregulation-GGPPS, CPS, KSL

Zhai et al. (2018)

Aspergillus fumigatusTS1,

Fusarium proliferatum BRL1

Oxalis corniculata

Indole acetic acid,

Gibberellins

P50-1,

P450-3,

P450-4,

ggs2,

des

Upregulation Bilal et al. (2018)
Aspergillus nidulans Rhizophora stylosa Abscisic acid

TBP,

TAF14,

XBP,

CDK7,

CCNH

Upregulation Xu et al. (2018)
Aspergillus niger CSR3 Cannabis sativa

Gibberellins,

Indoleacetic acid

P450-1,

P450-3,

P450-4,

ggs2,

des

Upgregulation-P450-4 Lubna et al. (2018)
Trichoderma atroviride D16 Salvia miltiorrhiza Tanshinones PHGPX Upregulation Peng et al. (2019)

Acetinobacter sp.

Marmoricola sp.

Papaver somniferum Benzylisoquinoline

T60D,

CODM,

COR

Upregulation by (SM3B)- T60DM, CODM, COR Ray et al. (2019)

Culvularia sp.,

Choanphora influndibulifera,

Aspergillus japonicus

Cathranthus roseus

Ajmalicine,

Serpentine

ORACA3,

ZCTs

Upregulation-ORACA3

Downregulation-ZCTs

Singh et al. (2020b)
Penicillium chrysogeum Ficus elastica Meleagrin roqO Upregulation Ding et al. (2020)
Mucor circinelloides Salvia mitiorrhiza Tanshinone DF20 Chen et al. (2021b)
Pyricularia oryzae Oryza sativa

Melanin,

Tenuazonic acid,

Nectriapyroles,

Pyriculols

NRPS-PKS Motayama et al. (2021)
Cladosporium tenuisssimum DF11 Salvia mitiorrhiza Tanshinone

HMGR,

DXS,

DXR,

GGPPS,

CPS,

KSL,

CYP76AHI

Upregulation Chen et al. (2022)

HMGR- 3-Hydroxy-3-Methylglutaryl-CoA Reductase;CYP71A12-Cytochrome P450 71A12; CYP71A13-Cytochrome P450 71A13; CYP71B15- Bifunctional dihydrocamalexate synthase/camalexin synthase; DXR- 1-deoxy-D-xylulose 5-phosphate reductor isomerase; GPPS- geranylgeranyl diphosphate synthase; CPS-Capsule polysaccharide locus; KSL-Kaurene synthase-like; CLS-Cardiolipin synthase; STS- Steroid sulfatase; TMNT-; P6H-Protopine 6-hydroxylase; TDC- Tryptophan decarboxylase; STR-Short tandem repeat; 16OMT-Tabersonine 16-O-methyltransferase; D4H-Deacetoxyvindoline 4-hydroxylase; DAT- Dopamine transporter; ORAC3-; ZTCs-; PRX1-Parired related homeobox 1; T60DM-Thebaine 6-O-demethylase; CODM-Codeine O-demethylase; Afua_6g 12,040-; TPS21-Terpene synthase 21;TPS11-Trihalos phosphatase/synthase 11; ggs2-Geranylgeranyl pyrophosphate synthase2; des-Desmin; PHGPX-Glutathione peroxidase; COR-Cold regulated; roqO-Cytochrome P450 monooxygenase roqO; NRPS-Non ribosomal peptide synthetase; PKS-Polyketide synthase; CYP76AHI- Cytochrome P450 enzyme 76AH1

Biosynthetic gene clusters and secondary metabolite production in fungi

It has been reported that there are a set of defined genes regularly clustered together in specified regions of fungal genome referred to as biosynthetic gene clusters (BGCs) that mediates the production of a large number of secondary metabolites in medicinal plants as well as in fungi (Chakraborty and Chakraborty 2021).

The genes located in these BGCs regions encode for various structural parts of the enzyme(s), and transport genes that take part in the biosynthesis of secondary metabolites that are co-regulated. Additionally, these genes are responsible for maintaining the operational integrity of various transcription factors (TFs) involved in the secondary metabolite production pathway (Fig. 2). Interestingly, the physical clustering of these BGCs augments the expression and regulation of genes involved in the formation of various substrates, and transport products, as well as alter the chemical scaffold of enzymes of key metabolic pathways of secondary metabolite synthesis (Kwon et al. 2021) (Fig. 2). Broadly fungal BGCs include genes for the synthesis of core enzymes like polyketide synthases (PKSs), non-ribosomal peptide synthetases (NRPSs), hybrids (PKS-NRPS), terpene synthases (TPS), and prenyltransferases (PTs) that alone or in combination directs the synthesis of the first product of biosynthesis pathway of fungal secondary metabolites (Avalos and Limón 2021; Sagita et al. 2021) (Fig. 2). Additionally, Sagita et al. (2021) showed that phenotype of secondary metabolite from endophyte fungi can be successfully linked to genotype by performing various experimental tools. Extensive variation has been seen in relation of the number of BGCs for different secondary metabolites among different fungal orders like Ascomycota, Basidiomycota, etc. (Mózsik et al. 2021).

Fig. 2.

Fig. 2

Global regulatory network for augmenting endophytic fungi mediated bioactive secondary metabolite production. Biosynthetic gene clusters (BGCs) are responsible for the production of enzymes, transcription factors, and transport genes involved in major metabolic biosynthesis pathways. These BGCs can be studied and analyzed through both conventional biology approaches as well as synthetic biology approaches. [Polyketide synthases (PKSs); non-ribosomal peptide synthetases (NRPSs); hybrids (PKS-NRPS), terpene synthases (TPS); prenyltransferases (PTs); one strain many compounds (OSMAC)]

A study by Kjærbølling et al. (2018) demonstrated the presence of a secondary metabolite cluster for aflatoxin, chlorflavonin, and ochrindol that are linked to three different Aspergillus species, i.e., A. ochraceoroseus, A. campestris, and A. steynii. Simultaneously, work also revealed clusters for secondary metabolite novofumigatonin, ent-cycloechinulin, and epi-aszonalenins in A. novofumigatus. Concomitantly, there are specific BGCs accountable for particular bioactive metabolites like cyclosporin produced by the fungus Tolypocladium inflatum has been shown to be encoded by 14 BGCs (Bushley et al. 2013). Metabolic survey of an endophytic fungus Calcarisporium arbuscula NRRL 3705 showed the presence of 65 BGCs responsible for the production of secondary metabolites like sordarin, aculeacin A, citreoviridin, alternariol, copalyl_diphosphate, etc. Among them, 23 gene clusters were encoding for PKS genes, 12 gene clusters of (NRPSs) along with some terpenes, and the remaining were PKS/NRPS hybrids gene clusters. Cheng et al. (2020) showed that the key secondary metabolite compound aurovertin, which is a polyketide-derived structure was mainly composed of seven genes, including aurA (these genes encode a PKS), aurB (a SAM-dependent methyltransferase), aurC (a FAD-dependent monooxygenase), aurD, aurE, aurF, and aurG (an acetyltransferase). In the fungus, Aspergillus flavus the BGCs comprise of hybrid PKS-NRPS cluster necessary for the synthesis of secondary metabolites 2-pyridones and leporins (Cary et al. 2015). Likewise, Wang et al. (2015a) reported the involvement of 27 PKSs, 12 NRPSs, five dimethylallyl tryptophan synthases, 15 terpenoid synthases, seven terpenoid cyclases, seven fatty-acid synthases, and five hybrids of PKS-NRPS for the synthesis of secondary metabolites in endophytic fungus Pestalotiopsis fici. In a different study, gene cluster encoding for two polyketide synthase enzyme (VdtA and VdtG) were identified in Paecilomyces variotii and Aspergillus viridinutans that are involved in biosynthesis of anti-bacterial compound viriditoxin (Urquhart et al. 2019). Whole-genome transcriptomic analysis of endophytic fungi, Alternaria sp. SPS-2 isolated from Echrysantha chrysantha showed that the protein-coding genes harbored 22 secondary metabolites BGCs including 7 PKSs, 10 NRPSs, 4 Terpenes, and 1 fungal-RiPP, and 14 of the BGCs are unknown (Tao et al. 2022). Recently, Yuan et al. (2022) identified 39 BGCs linked to 208 strains of A. oryzae using high end genome mining tools for 185 distinct terpenoid biosynthesis. Some of the examples of BGCs that have been studied in different endophytic fungi have been listed in Table 4. These BGCs mimic the gene clusters present in plants for the production of specialized metabolites (Polturak and Osbourn 2021). Due to recent advancement in genomics and other related tools, predicting the BGCs in endophytic fungi has become somewhat easy and as BGCse code of similar kind of secondary metabolites as that of plants, research efforts are being made to isolate secondary metabolites from endophytic fungi.

Table 4.

List of recent studies carried out for the identification of biosynthetic gene clusters (BGCs) in relation to bioactive secondary metabolite

Fungal strains Secondary metabolites Biosynthetic gene clusters References
Penicillium decumbens, Aspergillus aculeatus, Aspergillus versicolor Azaterrilone A PKS Grijseels et al. (2018)
Aspergillus flavus Aspergillic acid NRPS Lebar et al. (2018)
Colletotrichum gloeosporioides Cg01, Penicillium polonicum hy4 Huperzine A Epigenetic modification by histone methylation (HMTs), histone acetyltransferases (HATs), and histone deacetylase (HDACs) Kang et al. (2019)
Cochliobolus lunatus (TA26-46) Cochliobopyrones A and B, along with three isocoumarins and one chromone Chemical epigenetic manipulation by DNA methyltransferase inhibitor 5-azacytidine Wu et al. (2019)
Calcarisporium arbuscula NRRL 3705 Predominant metabolite aurovertins 65 BGCs including 23 PKS, 12 NRPS, 11 terpenes, seven PKS/NRPS hybrids,11 indoles and one other types Cheng et al. 2020
Aspergillus terreus Azaphilones Four BGCs, two nonreducing PKSs, one highly reducing PKS, and one NRPS-like Huang et al. (2020)
Penicillium brasilianum Brasiliamide A Epigenetic modulators such as suberoylanilide hydroxamic acid (SAHA) and nicotinamide (NAA) responsible for HDAC inhibition Akiyama et al. (2020)
Grammothele lineata SDL-CO-2015-1 Paclitaxel, taxol One NRPS, six Type I PKSs, 12 terpene and 10 NRPS-like Ehsan et al. (2020)
Aspergillus calidoustus, Aspergillus westerdijkiae Emericellamide A, emericellamide B, phenylahistin Epigenetic modification through histone deacetylase (HDAC) inhibition by vorinostat Aldholmi et al. (2020)
Penicillium dangeardii Azaphilones, Dangelone A-F, Dangelone G, Dangelone H, Dangeloside A-B, Didangelone A-E, Didangelone H, Didangelone F, Didangelone G, Tridangelone A-E 43 BGCs including 22 PKSs, eight NRPSs, eight PKSs/NRPSs hybrids, and five terpene synthases Wei et al. (2021)
Alternaria sp. SPS-2 Equisetin, Betaenones A–C, Alternariol, Dimethylcoprogen, Melanin 22 BGCs including 10 NRPSs, seven PKSs, four terpenes, and one fungal-RiPP Tao et al. (2022)
Helotiales sp. BL73 Cochlioquinone B and D, Isofusidienol A 77 BGCs including 26 PKS type I, three PKS type III, 25 NRPSs, six PKS/NRPS hybrids, and 13 terpene and four indole gene clusters Oberhofer et al. (2022)
Alternaria dauci Aldaulactone NRPS, PKS Courtial et al. (2022)
Sarocladium zeae Pyrrocidine PKS-NRPS hybrid Liu et al. (2022)
A. oryzae 185 distinct terpenoids 39 BGCs linked to 208 strains of A. oryzae Yuan et al. (2022)

Regulation of biosynthetic gene clusters by different stimuli and secondary metabolite production

Secondary metabolites produced by endophytic fungi are often regulated by stimuli that switch on inactivated BGCs of fungi and stimulate the production of particular bioactive metabolites (Mózsik et al. 2022) (Fig. 2). Additionally, some studies demonstrated activation of similar BGCs by different stimuli for various unrelated secondary metabolites thereby hinting towards the existence of cross-talk of multiple gene clusters. There are several approaches to switch on the multiple silent gene clusters responsible for encoding secondary metabolites. Stroe et al. (2020) showed that bacteria Streptomyces rapamycinicus during its interaction with the fungus Aspergillus fumigatus activates its previously silent BGCs and stimulate the production of fumigermin encoded by gene FgnA (a PKS synthase). This fumigermin has been shown to inhibit spores of bacteria and provide a competitive advantage to fungi. Interestingly, the production of secondary metabolites in fungi is extensively transcriptionally regulated and is being activated by different methods (Mózsik et al. 2022).

El Hajj Assaf et al. (2020) deciphered in their review that the production of secondary metabolite in Penicillium is regulated by various transcription factors as well as environmental stimuli. Transcription factors like basic leucine zipper (bZIP) are well known for their role in the regulation of secondary metabolite production. Endophytic fungus Pestalotiopsis fici CGMCC3.15140 present bZIP transcription factor PfZipA that regulates the production of isosulochrin, RES1214-1, and pestheic acid that are resistant to the oxidative reagents tert-butylhydroperoxide (tBOOH), diamide, and menadione sodium bisulfite (MSB) (Wang et al. 2015b). Various other transcription factors like Zn(II)2Cys6, Cys2His2, helix turn helix also regulate the production of key secondary metabolite in Aspergillus nidulans, Collettrichum lagenarium, and Acremonium chrysogenum (Rashmi and Venkateswara 2019).

Transcript expression of BGCs is often controlled by epigenetic regulation mediated by enzymes such as histone deacetylases (HDACs) and histone acetyltransferases (HATs) which are responsible for eliminating, establishing, silencing biosynthetic gene clusters, thereby resulting in the production or enhancement of a variety of secondary metabolites (Cichewicz 2010; Albright et al. 2015). A group of scientists, Magotra et al. (2017) reported in their study that epigenetic modifiers like valproic acid-induced tenfold enhancement of the production of secondary metabolites fumiquinazoline C in an endophytic fungus Aspergillus fumigatus (GA‑L7) isolated from Grewia asiatica. Various genes including Afua_6g 12040, Afua_6g 12050, Afua_6g 12060, Afua_6g 12070, and Afua_6g 12080 were involved in the biosynthesis of fumiquinazoline C and these were upregulated in the presence of epigenetic modifier, valproic acid. Further evidence of differential expression has been recently reported by Qadri et al. (2017), where small-molecule epigenetic modifiers induce the expression of secondary metabolism-related genes from an endophytic fungus, Muscodor yucatanensis Ni30. The researchers observed that in the Ni30 strain, small-molecule epigenetic modifiers such as suberoylanilide hydroxamic acid (SAHA) and 5-azacytidine were used for the over-expression of PKS genes. In this regard, bioactive extrolite brefeldin A was isolated from the wild variant of Ni30, and the production of ergosterol and xylaguaianol C was done from the epigenetic variant of endophytic fungus M. yucatanensis EV1.

Modern approaches for enhancing secondary metabolite production and analyzing biosynthetic gene clusters

Synthetic biology has significantly provided a new era of research in the production of secondary metabolite from endophytic fungi by manipulating BGCs (Fig. 2). Baral et al. (2018) comprehensively explained the various advanced methods like pleiotropic approaches, targeted genome-wide and, BGCs specific approaches that can be adopted to improve secondary metabolite production through endophytic fungi. Retro biosynthesis has also emerged one of the interesting way to for augmenting secondary metabolite production by exploring enzymes of particular secondary metabolite through studying enzymes of known s biosynthetic pathway of structurally similar secondary metabolites (Sagita et al 2021). One of the most common and long terms followed approaches that have been extensively utilized for producing a wide range of secondary metabolites is the OSMAC approach, i.e., “one strain many compounds” in which cultural conditions are being manipulated like aeration, light, temperature, etc. for the isolation of novel secondary metabolites (Fig. 2) (Ramírez-Villalobos et al. 2023; Staropoli et al. 2023) Remarkably, previous literature has also shown that certain chemical inducers and environmental cues also stimulate the production of secondary metabolites (Xue et al. 2023) Apart from this, co-cultivation using different bacteria or different microbial partners also stimulates the production of fungal metabolites (Akone et al. 2016; Vinale et al. 2017a; Wakefield et al. 2017; Abdelwahab et al. 2018; Xue et al. 2023).

It has been established that co-culture with microbes for the augmentation of plant secondary metabolite concentration is very momentous approach and will drastically improve secondary metabolite composition of plants (Zhi-lin et al. 2007). These fungal endophytes acts as a elicitor molecule during co-culture for escalating the production of secondary metabolite in plants (Hussain et al. 2021a, b). A study by Ding et al. (2018) demonstrated significantly improved bioactive secondary metabolite production in tissue cultured Rumex gmelini seedlings when co-cultured with endophyte Aspergillus sp. An interesting research by Li et al. (2009) demonstrated increased production of important anti-cancerous drug paclitaxel in a co-bioreactor where cell suspension cultures of Taxus plant and an endophytic fungi Fusarium mairei were co-cultured. It has been observed that co-culture to cell suspension with endophytic fungus led to increased activity of enzymes responsible for production of secondary metabolites as evident from the work of Tang et al. (2011) where activity of enzymes phenylalanine ammonia lyase (PAL) and TDC were found to be increased in cell suspension cultures of Cathranthus roseus on inoculation with Fusarium oxysporum under in-vitro conditions. Increase biosynthesis of tanshinone was observed in seedlings of medicinal plant Salvia miltiorrhiza root co-cultured with an endophytic fungus Mucor circinelloides DF20 (Chen et al. 2021a, b, c). Similar, beneficial effects of co-culturing of endophytic fungi for improving secondary metabolite concentration were seen in the recent work of Devi et al. (2023b) where inoculation of Stevia rebaudiana with endophytic fungi Fusarium fujikuroi promoted substantial increase of polyphenols and rutin in comparison to control plants without any inoculation. Overall fungal endophytes has great potential in regulating the production of secondary metabolite concentration in plants and elicitation is one of the major approach that has tremendous benefit for increasing large-scale production of secondary metabolite during cell or plant culture (Isah et al. 2018). One of the major advantage of using fungal endophytes for the extraction of secondary metabolite is the yield and it was observed that there eightfold increase in taxol production by the endophytic fungus Paraconiothyrium SSM001 when co-cultured with Alternaria and Phomopsis isolated from the bark of the same host plant (Soliman and Raizada 2013a, b). Similarly, Bhalkar et al. (2016) demonstrated that co-culture of two endophytic fungi isolated from Nothapodytes nimmoniana Colletotrichum fructicola SUK1 and Corynespora cassiicola 25 SUK2 escalated the production of camptothecine an important anti-cancer drug. The study revealed that using response surface methodology (RSM) and mixed fermentation with two fungi there was increased production of camptothecine (146 mg l−1) in comparison to their monocultures (Bhalkar et al. 2016).

Several other methodologies that can be regulated for the extraction of novel secondary metabolites from fungi are heterologous expression, chromatin remodeling, cluster-specific regulation, etc. With the advent of genomic advancement, target specific, i.e., promoter modification of BGCs has been carried out to augment secondary metabolite production. Keller (2019) depicted the presence of an extensive number of BGCs that may account for only a few secondary metabolites, although there are a plethora of BGCs that are still being explored and may lead way to the discovery of new bioactive metabolites that might have application in agriculture, pharmaceutical, etc.

Kjærbølling et al. (2019) reviewed the application of various molecular and genomic tools for understanding these BGCs and secondary metabolite production. Next-generation sequencing has played a pivotal role in the identification and correlation of BGCs with different secondary metabolite production among different fungal species as reviewed by Cacho et al. (2015). By analyzing these BGCs, biosynthesis of novel metabolites useful for human and animal health can be carried out as well as production of important metabolites responsible for providing defense to plants against various abiotic and biotic stress can be strategically enhanced using genetic manipulation techniques (Fig. 2). Subsequently, Ren et al. (2020) also reviewed the application of various bioinformatics tools such as AntiSMASH, PlantiSMASH, NP.searcher, SMURF, ClustScan, eSNaPD, Cluster Finder, EvoMining for the identification of BGCs responsible for the production of bioactive natural product in different plant and microbes (Fig. 2). By analyzing these BGCs, one can predict chemo diversity based evolutionary lineage among fungi as reviewed by Rokas et al. (2020). Theobald et al. (2018) carried out extensive genome mining using SMURF and antiSMASH for the identification of BGCs of Aspergillus section Nigri responsible for the malformin biosynthesis, an important secondary metabolite. Simultaneously, phylogenetic studies were completed to categorize different Aspergillus species based on BGCs of malformin biosynthesis in different clades. Deep learning has also been emerged as a new technology to understand the endophyte and bioactive secondary metabolite production gene network and bioprospection a evident from the study of Aghdam and Brown (2021)

Endophytic fungi mediated gene expression for growth, nutrient acquisition, and defense

Endophytic fungi in various plants augment plant biomass, seed production, and tolerance against different abiotic and biotic stresses (Vahabi et al. 2013; Toppo et al. 2022). Many studies have shown the positive effects of endophytic fungi on plant growth (Baron and Rigobelo 2022), nutrient acquisition (Verma et al. 2021), and stress alleviation (Karimi et al. 2022) due to which these endophytic fungi have now been utilized as biostimulants in modern agriculture under climate change conditions. Concomitantly, molecular studies have confirmed that these endophytic fungi affect the expression of various genes involved in various growth and metabolic pathways as well as defense (Rauf et al. 2022). The colonization of the endophytic fungus Piriformospora indica in the roots of trifoliate orange manifested effective results on plant growth and nutrient accession, especially P. There was up-regulation in the expression of the PtPT3, PtPT5, PtPT6 genes mainly in the epidermal cells and outer cortex of the root by a single inoculation of the endophytic fungus P. indica resulting in the better uptake of P at the root-soil configuration (Yang et al. 2021). Endophytic fungus Bipolaris sp. CSl-1 when inoculated in the soybean plant remarkably improves the root length, shoot and root fresh and dry weight, chlorophyll content, and concentration of SA under NaCl stress (Lubna et al. 2022). At the same time, inoculation of Bipolaris sp. CSl-1 enhanced soybean resistance towards NaCl stress and down-regulated the expression of the genes GmFDL19, GmNARK, and GmSIN1 and intensifies plant growth under salt stress (Lubna et al. 2022).

A study by Camehl et al. (2011) showed that growth promotion by Piriformospora indica in Arabidopsis was mediated by gene (OXI1) (Oxidative Signal Inducible 1). It was seen that P. indica upregulated transcript expression of OXI1 and PDK1 while downregulated the expression of the defense genes. In a different study, inoculation of endophytic fungus Fusarium oxysporum strain F047 induced resistance against Fusarium wilt in tomato by increasing transcript expression of three genes CH31, GLUA, and PR-1-a (Aimé et al. 2013). Endophytic fungus Trichoderma asperelloides T203 colonized in the roots of the plant Arabidopsis thaliana enhanced the expression level of the genes WRKY18 and WRKY40, which energized the expression of the defense genes FM01, PAD3 and CYP1A13. The wrky18/wrky40 double mutant line had reduced root colonization while over-expression of WRKY40 resulted in the partial phenotypic complexion. It has been observed that Trichoderma inoculated plants increased the expression of MDAR gene resulting in plant growth, osmo-protection as well as tolerance towards general oxidative stress in the root of the plant (Brotman et al. 2013). Defense-related genes were discriminatively expressed on the tomato plants colonized with the endophytic fungus Fusarium solani strain K (FsK) after spider mite affliction in comparison to those of the spider mite-infested un-colonized plants. The genes GLU-A and CHI9 were predominantly induced against the pathogens that were upregulated in the FsK-colonized plant. Substantially, upregulation of the genes PI-IIc and PR-1A and downregulation of the gene PPO-F occurred on the FsK-colonized plants. Furthermore, a remarkable further increase in the transcript levels of WIPI-II was led by FsK-colonization (Pappas et al 2018). Rondot and Reineke (2019) demonstrated upregulation in transcript expression of genes PR-2 (β-1,3-glucanases), PR-3 (chitinases), PR-5 (thaumatin-like proteins), and the PR protein 10.3 with inoculation of the endophytic fungus Beauveria bassiana against the infections by grapevine downy mildew Plasmopara viticola. Additionally the study also showed genes related to the stilbene synthesis and other related genes that were also upregulated that were involved in the plant pathogen interaction and defense of grapevine against the pathogen (Rondot and Reineke 2019). The genes LOX1 and OPR7 in the JA biosynthesis were upregulated by the inoculation of the endophytic fungus Metarhizium robertsii in the maize plant and led to improved plant defense mechanisms (Ahmad et al. 2020). Similarly, in the SA response pathway, PR5 was upregulated along with chitinase gene ECH A, whereas, the PR4 was downregulated in the leaf tissue isolated from the plants grown from M. robertsii -inoculated seeds (Ahmad et al. 2020). The wheat spikes treated by the endophytic fungus Penicillium oslonii ML37 showed faster and stronger expression of the defense metabolism against the Fusarium head blight in wheat (caused by Fusarium graminearum). The transcriptional activation of WRKY transcriptional factors was seen as a response to the fungal colonization which lowered the metabolites 15-acetyl-DON and culmorin of the pathogen F. graminearum (Rojas et al. 2022).

Emerging applications of endophytic fungi in different biotechnological sectors

Novel bioactive compounds produced by various endophytic fungi are of biotechnological attention as they can be potentially utilized as a source of secondary metabolites, anti-microbial agents, biological control agents, immune-suppressant, antiviral compounds, anti-tumor compounds, anti-diabetic, antihypertensive, anti-inflammatory, anti-atherogenic, anti-thrombotic, cardioprotective, vasodilatory and anti-allergic agents, antibiotics, a natural anti-oxidant, and much more (Yadav 2018; Yan et al. 2018). Thus, bioactive compounds from endophytic fungi can principally be exploited in multifaceted arenas, ranging from biomedicine, pharmaceutical drugs, agriculture, and industrial uses to biofuel and biocatalytic developments (Liu et al. 2017; Pan et al. 2017). In this direction, a prospective strategy can be to develop strain improvement methods using genetic engineering, optimization of culture conditions/media for cultivation, and co-culturing of different endophytic strains. Recent advances in high-throughput technologies and the “genomic revolution” have contributed considerably to natural product research and the discovery of biosynthetic gene clusters (Zazopoulos et al. 2003; Lautru et al. 2005). Genetic engineering of endophytes is still in its infancy and is often regarded as a progenitor of system biology and functional genomics strategies (Tyo et al. 2007) with the potential for long-term translational success. Studies have suggested the inclusion of metabolic pathways and genes in endophytes via genetic recombination between plant hosts and endophytes. In addition, horizontal gene transfer, an evolutionary mechanism, has been suggested as an adaptive mechanism for endophytes, and it confers novel traits to the associated microbes (Kumari et al 2016; Tiwari and Bae 2020).

Recently, harnessing microbial resources via biotechnological means has been promising; with vast applications in agricultural and environmental studies. Current research on plant growth-promoting endophytic fungi has revealed more insights into their biotechnological importance in the synthesis of biofertilizers and biopesticides in developing agriculturally friendly chemicals to ensure maximum food security in a sustainable and safe manner (Igiehon and Babalola 2017). The culture-dependent approach has been used to isolate various microorganisms from different environments, however only a small proportion of total microbes can be cultured; hence, the large microbial population can be assessed using culture-independent metagenomics approaches (focused on gene sequencing, and further computational analysis of sequence data from microbial DNA to detect structural, functional, and metabolic pathways) (Dissanayake et al. 2018; Fadiji and Babalola 2020).

Metagenomic analysis of endophytes from various plant organs allows to determine the structural, functional, and phylogenetic construction of genetic relatedness (from a metagenomic library) in the microbial genome from long reads of metagenome sequence data. Next-generation sequencing approaches such as 454 pyrosequencing (Roche) and Illumina sequencing have been employed in the study of whole microbial communities with novel traits responsible for growth promotion, cellular metabolism, phytohormone synthesis, and nitrogen fixation from the internal tissues of plant roots. At the gene level, the analysis of gene prediction, taxonomic profiling, and molecular functions of plant growth-promoting microorganisms are permissible using metagenomic analysis (Hardoim et al. 2015).

Recently, in-vitro preparation of the Cas9-gRNA ribonucleoprotein complex and transformation into protoplasts of filamentous fungi has become available (Pohl et al. 2016; Al Abdallah et al. 2017; Wang et al. 2018). Although it has not been applied in endophytic fungi, this is a promising strategy to advance not only in gene editing, but it also advances the development of gene expression for biotechnological purposes using the recently developed CRISPR interference and CRISPR activation systems which offer novel strategies to regulate gene expression (Zheng et al. 2019). To a large extent, the use of omics technology has advanced microbial studies to a level of genomics, proteomics, and transcriptomics (Akinola and Babalola 2020). However, there are challenges like diffusion of plant DNA compared to microbial DNA (making it difficult to separate fungal metagenome), the inability to manage and conduct automation classification of large sequence datasets, and the unattainability of functional gene annotations in the gene library for fungal genomes under examination. Due to the abundance of plant DNA compared to fungal DNA, it is difficult to successfully sequence only microbial DNA at high coverage. Very recently, the enrichment of fungal DNA from the stems and leaves of maize, rice, and soybean (Mehmood et al. 2019) and the internal tissue of potato tubers has been reported (Krell et al. 2018). With the current ongoing metagenomics research of the microbial world, scientists could efficiently engineer fungal endophytes and harness their possible biological resources to formulate bio-inoculants in promoting plant growth for improved agricultural productivity.

Biocontrol agents

Extensive and irrational usage of chemical pesticides to regulate phytopathogens has caused environmental deterioration, residue complications, resistance to pathogens, and diverse threats to the health of living organisms (Liu et al. 2016). However, various bioactive secondary metabolites being produced by endophytic fungi act as biocontrol agents and have been extensively described to help hosts by improving resistance to pathogenic invaders (Yu et al. 2017). Based on this, few studies on efficient endophytes as the biocontrol agents of phytopathogens have been illustrated in Table 5. Endophytic fungi can suppress pathogens by inducing morphological changes and overexpression of endogenic anti-microbial compounds in hosts, competition for niche utilization, anti-biotics production, egg parasitic (breakdown of the shell integrity), hatching inhibition, (Ownley et al. 2008; Siddaiah et al. 2017). Besides providing resistance to stresses, these fungi can also augment plant development by stimulating nutrient availability (e.g. solubilizing different sources of phosphorus, improving plant–endophyte–herbivore communications for nitrogen cycling), and transferring rhizospheric nutrients to their plant hosts, by decomposing soil organic compounds and/or by stimulating growth hormones like indole acetic acid in host plants (Kiers et al. 2011; Priyadharsini and Muthukumar 2017). Fungal-derived polysaccharides especially exopolysaccharides may aid these fungi to resist various abiotic stresses, assist in the establishment of biofilms for colonizing the host, turn into bio-elicitors to stimulate efficient metabolite to build up in plant partners, and/or employ many other biological activities (Liu et al. 2017). These microbial polysaccharides can be applied for the production of many bioproducts, e.g., coverings in wound shock healing, suspension stabilizers, tablet granulation tissue and coating, and many more (Freitas et al. 2011; Moscovici 2015).

Table 5.

A list of endophytic fungi with application in various other biotechnology sectors

Name of endophyte Host plant Consequence Reference
Biocontrol agent

 Fusarium solani TRX-34–1,

 Rhexocercosporidium sp.

Sophora tonkinensis Gapnep Biocontrol agents against phytopathogens of Panax notoginseng Yao et al. (2017)
 Fusarium verticillioides Zea mays Biocontrol—Restrict Ustilago maydis growth Rodriguez et al. (2012)

 Penicillium sp.,

 Guignardia mangiferae,

 Hypocrea sp.,

 Neurospora sp.,

 Eupenicillium javanicum, Lasiodiplodia theobromae

Cucumis sativus Biocontrol of Fusarium oxysporum f. sp. cucumerinum Abro et al. (2019)
 Alternaria alternata Grapevine leaves

Production of three anti-fungal metabolites

• Three diketopiperazines: cyclo (l-phenylalanine-trans-4-hydroxy-l-proline)

• Cyclo (l-leucine-trans4-hydroxy-l-proline)

• Cyclo (l-alanine-trans-4-hydroxy-l-proline)

Effective against Plasmopara viticola

Musetti et al. (2006)
 Cryptosporiopsis sp. and Phialocephala sphareoides Norway spruce (Picea abies) Inhibit phytopathogen (anti-fungal)—Heterobasidion parviporum, Phytophtora pini, and Botrytis cinerea Terhonen et al. (2016)
 Root-knot nematode (Meloidogyne spp.) Roots of almost all cultivated plants Biocontrol effect against Meloidogyne incognita Yao et al. (2015)
 Xylaria Pinus strobus Production of secondary metabolite including terpenoids, cytochalasins, mellein, alkaloids, and polyketides—promising herbicide, biopesticide, and biocontrol agents Macías-Rubalcava and Sánchez-Fernández (2017)
 Fusarium proliferatum

Macleaya cordata (plume poppy), Sanguinaria canadensis (bloodroot), and

Chelidonium majus (swallowwort poppy)

Sanguinarine—Anti-bacterial

Anti-inflammatory

Anti-helminthic

Wang et al. (2014)
 Guignardia mangiferae Citrus sp.

Can synthesize 5–30-nm-sized, spherical-shaped AgNps of Anti-bacterial

Anti-fungal and Anti-proliferative activity

Balakumaran et al. (2015)
 Pestalotia sp.

Leaves of Syzygium

cumini

AgNps with anti-microbial activity can be increased by combination with antibiotics such as gentamycin and sulphamethizole. Together they can inhibit the growth of the human pathogens Staphylococcus aureus and Salmonella typhi Raheman et al. (2011)
 Eupenicillium parvum Azadirachta indica Azadirachtin A and B and their structural analogs—bioinsecticides with antifeedant and insect growth-regulating properties Kusari et al. (2012)

 Guignardia mangiferae,

 Fusarium proliferatum,

 Colletotrichum gloeosporioides

Taxus x media Taxol—Fungicide Xiong et al. (2013)
Stress tolerance/ Bioremediation/Biosorption
 Piriformospora indica Hordeum vulgare, Brassica rapa Biotic/abiotic stress tolerance especially Drought stress tolerance Sun et al. (2010), Johnson et al. (2013) and Ghaffari et al. (2019)
 Mucor sp. MHR-7 Brassica campestris Metal toxicity reduction Zahoor et al. (2017)

 Rhizopus sp. CUC23, A. fumigatus ML43

 Penicillum radicum PL17

Lactuca sativa Chromium detoxification Bibi et al. (2018)
 Neotyphodium coenophialum, N. uncinatum

Festuca arundinacea,

Festuca pratensis

Bioaugmentation, total petroleum hydrocarbons (TPH) and polycyclic aromatic hydrocarbons (PAHs) removal from the soil Soleimani et al. (2010)
 Verticillium sp., Xylaria sp.

Plants from

Ecuadorian Amazon

Degradation of Petroleum hydrocarbon Marín et al. (2018)
 Curvularia sp., Neusartorya sp. Mangrove sp. Heavy metal biosorption Onn et al. (2016)
 Bjerkandera adusta SWUSI4 Sinosenecio oldhamianus Detoxification of triphenylmethane dyes Gao et al. (2020)
 Lasiodiplodia theobromae Boswellia ovalifoliolata Heavy metal tolerance Aishwarya et al. (2017)
 Lindgomycetaceae P87, Aspergillus sp. A31 Aeschynomene fluminensis Heavy metal resistance, bioremediation Pietro-Souza et al. (2020)

 An endophytic fungus,

 Lasiodiplodia sp. MXSF31

Stem of

Portulaca oleracea

Fungi have the potential in promoting the growth of Portulaca in metal-contaminated soils. The biosorption process of MXSF31 was attributed to the functional groups of hydroxyl, amino, carbonyl, and benzene ring on the cell wall Deng et al. (2014)
 Fusarium sp. Mangroves Tailored Biosorption—Endophytic fungi were dried and powdered and then chemically modified by formaldehyde, methanol, and acetic acid to augment their affinity for uranium from polluted wastewater Chen et al. (2014)
Plant growth promotion/Biofertilizers

 Penicillium sp. 21,

 Penicillium sp. 2,

 Aspergillus sp. MNF

Camellia sinensis Mineral-solubilizing function (Ca3(PO4)2 and rock phosphate) Gupta et al. (2007) and Nath et al. (2012)
 Trichoderma gamsii (NFCCI 2177) Lens esculenta Solubilization of Tricalcium phosphate Rinu et al. (2014)
 Trichoderma peudokoningi, Chaetomium globosum, Fusarium oxysporum Solanum lycopersicum Siderophore production, HCN and ammonia production Neha et al. (2015)
 Ophiosphaerella sp., Cochliobolus sp. Triticum aestivum PGP activities Spagnoletti et al. (2017)
 Cladosporium sphaerospermum Glycine max Solubilize calcium phosphate Hamayun et al. (2009)
 Fusarium tricinctum RSF-4L, Alternaria alternata RSF-6L S. nigrum Production of phytohormones (gibberellins) Khan et al. (2015)

 Coniothyrium aleuritis 42, Pichia guilliermondii F15, Fusarium oxysporum NSF2,

 F. proliferatum AF04, Aspergillus nidulans FH5, Trichoderma spirale YIMPH30310

Lycopersicon esculentum Plant biomass increase, fruit yield Xia et al. (2019)
 Phialocephala fortinii Chamaecyparis obtuse, Rubus sp. Promote Asparagus officinalis growth Surono and Narisawa (2017)
 Phomopsis liquidambari Arachis hypogaea Increase nodulation and N2 fixation by enhancing hydrogen peroxide and nitric oxide signaling Xie et al. (2017)
 Metarhizium robertsii

Phaseolus vulgaris

Glycine max

Panicum virgatum

Triticum aestivum

The insect-pathogenic fungus

Metarhizium robertsii couple root association and insect (Galleria mellonella) pathogenicity so that it acts as a conduit to provide insect-derived nitrogen to plant hosts

Behie and Bidochka (2014a, b)
 Penicillium chrysogenum Pc_25 Alternaria alternata Aa_27 Asclepias sinaica Produces ammonia, indole acetic acid (IAA), and phytohormone to improve the growth parameters of plant Fouda et al. (2015)

Alternaria sp.,

 Fusarium tricinctum RSF-4L, Alternaria alternata RSF-6L

Brassica napus, Crocus sativus Linn., Solanum nigrum Growth-promoting—Indole-3-acetic acid Shi et al. (2017), Wani et al. (2016) and Khan et al. (2017)

 Acremonium curvulum, Aspergillus niger,

 Cochliobolus lunatus,

 Gibberella baccata, Myrmecridium schul zeri, Myrothecium verrucaria,

 P. commune,

 Phoma putaminum,

 Pithomyces atro-olivaceus, Trichoderma piluliferum

Bauhinia Forficate for the production of cellulase, lipase, protease, and xylanase Bezerra et al. (2015)
Pharmaceutical industry
 Taxomyces andreanae, T. brevifolia Pacific yew Paclitaxel—Anti-cancer Stierle et al. (1993)
 Fusarium subglutinans Tripterygium wilfordii Subglutinol A—Immuno-suppressant Strobel and Pliam (1997)
 Trametes hirsuta Podophyllum hexandrum Podophyllotoxin—Antiviral, Radio-protective Puri et al. (2006)
 Rhizoctonia bataticola Coleus forskohlii Forskolin—Anti-HIV, Anti-tumor Mir et al. (2015)
 Fusarium proliferatum Macleaya cordata Sanguinarine—Antihelmintic Wang et al. (2014)
 Alternaria sp. Digitalis lanata Digoxin—Cardiotonic Kaul et al. (2013)
 Phomopsis sp. Cinchona ledgeriana Quinine—Antimalarial Maehara et al. (2010)
 Alternaria alternata Capsicum annuum Capsaicin—Cardio-protective Devari et al. (2014)
 Fusarium solani Rheum palmatum L Rhein—Anti-microbial, Anti-tumor and Anti-inflammatory You et al. (2013)

 Taxomyces andreanae, Cladosporium cladosporioides, Paraconiothyrium SSM001, Guignardia mangiferae,

 Fusarium proliferatum,

 Colletotrichum gloeosporioides

Phloem (inner bark) of the Pacific yew, Taxus brevifolia or Taxus media

Taxol, a diterpenoid anti-cancer drug, originally been extracted from Taxus brevifolia or Taxus media. Taxol-producing fungal endophytes, like Taxomyces andreanae, Cladosporium cladosporioides, Paraconiothyrium SSM001, Guignardia mangiferae,

Fusarium proliferatum,

Colletotrichum gloeosporioides are being actively developed as alternative means for the production of taxol

Zhang et al. (2009); Soliman and Raizada (2013a, b); Xiong et al. (2013) and Soliman et al. (2015)
 Chaetomium globosum Polysiphonia urceolata Chaetopyranin—benzaldehyde derivative isolated from fungal endophytes has anti-cancerous and anti-oxidant properties Wang et al. (2006)
 Pestalotiopsis fici Camellia sinensis Novel chromone Pestalotiopsone F 22 reported from the culture filtrate of fungal endophyte exhibit adequate cytotoxic effect against the murine cancer cell line L5178Y Xu et al. (2009) and Liu et al. (2009)
 Fusarium oxysporum Ephedra fasciculata Beauvericin—a depsipeptide isolated from the fungal endophyte repress movement of the metastatic prostate cancer (PC-3M) and breast cancer (MDA-MB-231) cells Zhan et al. (2007)

 Trichoderma atroviride, Fusarium solani,

 Fomitopsis sp.,

 Alternaria alternata,

 Phomopsis sp.

Catharanthus roseus acuminata,

Miquelia dentata

Camptothecin (CPT) is a monoterpenoid indole alkaloid and its derivatives (inhibit DNA topoisomerase I) are anti-cancer agents targeting small lung and refractory ovarian cancers. CPT and its derivatives were originally extracted from Camptotheca acuminata and Miquelia dentata Pu et al. (2013); Kusari et al. (2011) and Shweta et al. (2013)
 Endophyte Mimosops elengi Ergoflavin (anti-cancer compound), a pigment isolated from endophyte inhibit the production of TNF-α and IL-6 Deshmukh et al. (2009)

 Nigrospora sphaerica,

 Talaromyces radicus

Catharanthus roseus Vinca alkaloids (Vas) are naturally found in Catharanthus roseus. Well-known anti-cancer compounds (used in the treatment of leukemia, and Hodgkin’s disease) include vinorelbine, vindesine, vincristine, and vinblastine Ayob et al. (2017) and Palem et al. (2015)
 Penicillium oxalicum Gymnema sylvestre Gymnemagenin—Antidiabetic Parthasarathy and Sathiyabama (2014)
 F. oxysporum SYP0056 Ginkgo biloba produces Ginkgolide B, which used in the treatment of cardiovascular diseases Cui et al. (2012)
 Chloridium species Azadirachta indica Javanicin (naphthaquinone), with anti-microbial properties, extracted from endophytic fungus Kharwar et al. (2009)
 Chaetomium leaves of Sapium ellipticum Produce chaetocochin A-C having anti-cancer properties Akone et al. (2016)
 Exserohilum rostratum Amazon Plant (Bauhinia guianensis) Polyketide monocerin has antagonist activity against E. coli (ATCC 25922), Pseudomonas aeruginosa (ATCC 27853), Staphylococcus aureus (ATCC 25923), Bacillus subtilis (ATCC 6633), and Salmonella typhimurium (ATCC 14028) Pinheiro et al. (2017)
 Phomopsis sp. XP-8 Eucommia ulmoides Oliver Antihypertensive—Pinoresinol diglucoside, Pinoresinol monoglucoside, Pinoresinol Shi et al. (2012) and Yan et al. (2015)
 Fusarium solani SD5 Alstonia scholaris (devil tree) Produce PS-I (capsular polysaccharide) with anti-inflammatory activities Mahapatra and Banerjee (2012, 2013)
 Diaporthe sp. JF766998, Diaporthe sp. JF767007 Piper hispidum Sw Antiproliferative activity against human breast carcinoma (MCF7) and hepatocellular carcinoma (HepG2-C) cell Orlandelli et al. (2017)
 Penicillium chrysogenum MTCC 5108 Mangroves Source of non-ribosomal peptides (NRP) and Penicillin (antibiotic) Devi et al. (2012)
Anti-oxidants
 Pestalotiopsis microspora VJ1/VS1 Gymnema sylvestre AgNPs—Anti-oxidant and Anti-cancer Netala et al. (2016a)
 Mucorfragilis fresen, Eupenicillium parvum Sinopodophyllum hexandrum Kaempferol—Anti-oxidant and anti-microbial activity Huang et al. (2014)

 Fusarium sp. JZ-Z6,

 F. sp. JZ-Z7,

 F. redolens Sz1_1 H,

 F. tricinctum WS11790

Fritillaria unibracteata Anti-oxidants with anti-cancerous activity—Gallic acid, Phlorizin and Rutin Pan et al. (2017)

 Arthrinium sp. 0042,

 Colletotrichum sp. 0047/0048,

 Diaporthe sp. 0051

Aquilaria subintegra Anti-oxidants—oxo-Agarospirol, α-Agarofuran, β-Agarofuran, δ-Eudesmol and β-Dihydro agarofuran Monggoot et al. (2017)
 Sordariomycete sp. B5 Eucommia ulmoides Oliver (Du-Zhong)

Chlorogenic acid (phenolic ester of caffeic and quinic acids)—Anti-bacterial

Anti-oxidant, Anti-tumor and Anti-fungal

Chen et al. (2010)
 Berkleasmium sp. Dzf12 Dioscorea zingiberensis (yellow ginger) Produce Exopolysaccharide, with anti-oxidant activities Li et al. (2012a)
 Fusarium oxysporum Dzf17 Dioscorea zingiberensis Produce water-soluble polysaccharides (WPS) with anti-oxidant activities Li et al. (2011, 2012b)
 Aspergillus sp. Y16 Mangrove As1-1, an exopolysaccharide with anti-oxidant properties Chen et al. (2011)
 Aspergillus versicolor ENT7 Centella asiatica

AgNPs—Anti-bacterial, Anti-fungal and

Anti-oxidant activity

Netala et al. (2016b)
 Dabaryomyces hansenii The fermentation of black tea with endophytes has resulted in the addition of major vitamins such as A, B1, B2, B12, and C, which are sufficient to fulfill the recommended dietary allowance. It also resulted in the reduction of tannins and caffeine in an effective amount. Moreover, the compound theophylline, which accumulates due to fermentation, imparted a bronchodilatory effect to the tea Pasha and Reddy (2005)
Biofuels
 Gliocladium roseum NRRL 50072 Eucryphia cordifolia (ulmo) Produce a series of volatile hydrocarbons and their derivatives on oatmeal-based agar and cellulose-based media in a species-specific manner Strobel et al. (2008) and Griffin et al. (2010)

 Penicillium brasillianum, Penicillium griseoroseum,

 Xylaria sp. (NICl3),

 Xylaria sp. (NICL5),

 Penicillium sp. (PAOE),

 Trichoderma spp.

From several different species of tropical plants Produce high concentrations of a lipid matrix that may serve as promising sources of biofuel precursors Santosfo et al. (2011)
 Nigrograna mackinnonii Produces a series of volatile natural products, including terpenes and odd chain polyenes—a source of biofuel Shaw et al. (2015)
 Gliocladium sp. Eucryphia cordifolia (ulmo) Degrade plant cellulose and synthesize complex hydrocarbons under microaerophilic conditions. This fungus could produce hydrocarbons ranging from C6 to C19 (i.e., hexane, heptane, as well as benzene) directly from cellulosic biomass Ahamed and Ahring (2011)

 Hypoxylon sp.,

 Daldinia eschscholzii

Sources of biomass deconstructing carbohydrate-active enzymes (for conversion of lignocellulose into advanced biofuels) Wu et al. (2017)
 Periconia sp. Torreya grandifolia Produces thermotolerant β-glucosidase, a thermotolerant secondary metabolite which has high activity toward cellobiose and carboxymethylcellulose. The enzymes can convert lignocellulosic biomass into biofuels and chemicals Harnpicharnchai et al. (2009)

Bioremediation

Continued industrial development in the world is leading to the accelerated emission of various pollutants including heavy metals (HMs), anti-biotics, and pesticides (Ma et al. 2011; Zouiten et al. 2016; Vignaroli et al. 2018). These pollutants leak from industrial surpluses and aquatic resources into soils and cause environmental pollution, adversely affect plant growth and raise toxic food chain concerns (Deng and Cao 2017; Hassan et al. 2017). Remediation strategies available for reducing the damaging effects at HMs-contaminated sites include physicochemical approaches such as filtration treatment, excavation, on-site metal stabilization, and phytoremediation. All these strategies have their limitations (beyond the scope of this chapter), thus, recent research indicates that bioremediation like the utilization of endophytic fungi (a few studies mentioned in Table 5) has emerged as an eco-friendly technique to clean metal-contaminated areas (Xiao et al. 2010; Deng et al. 2014; Khan et al. 2017; Zahoor et al. 2017). The fungi can eliminate rhizospheric HMs by biotransformation, biosorption on the fungal cell wall, and/or their buildup in fungal hyphae, thus dropping their disposal and harmfulness to host plants (Khan et al. 2017; Zahoor et al. 2017). Since bio-adsorption efficiency can be improved by modulating the density of the functional groups on a fungal cell wall, endophytic fungi can be tailored into a significantly efficient biosorbent (with improved biosorption efficiency for metal ions) by making chemical modifications. Other aspects comprise interaction time, pH, solid/liquid ratio, and original HMs levels in the polluted areas (Chen et al. 2014). It was observed that endophyte fungus can also be exploited for the commercial production of enzymes as evident from studies by Toghueo et al. (2017) that showed remarkable production of extracellular enzymes such as cellulases, amylases, lipases, and laccases from fungal endophytes. Recently, Abdalla et al. (2020) also showed the production of cellulases and xylanases from the endophytic fungus isolated from medicinal plants of South Africa. The study depicted the presence of the polyketide synthase type 1 (PKS1) gene that will be utilized for the production of bioactive metabolites in future. Several endophytes isolated from the medicinal plant Kadsura angustifolia were found to have degradative activity (Huang et al. 2015).

Secondary metabolite

Plants are a potential synthesizer of functional secondary metabolites—the source of crude natural drugs (Venugopalan and Srivastava 2015), but dependency on the plant growth rate, little quantities of drug yield/plant, laborious extraction, and over-harvesting lead to extinction distress in some rare medicinal plants (Huang et al. 2014) make plants inappropriate source of drugs. In this direction, many studies have provided key insights into the existing and emerging significance of fungal endophytes in drug discovery and research (Tiwari and Bae 2022). In contrast, endophytic microorganisms also exhibit competence to yield associated plant secondary metabolites with therapeutic and cosmetics values (Budhiraja et al. 2013; Kaushik et al. 2014). Cultivation of such fungal endophytes has been exploited as more efficient large-scale production systems for valued metabolites as they are fast-growing with reduced space utilization, and can be easily modified as per requirement (Palem et al. 2015).

The rising mandate for fuels and ecological complications triggered by the emission of greenhouse gases have directed towards a convincing necessity of sustainable alternate energy sources like biological organisms (Wu et al. 2017). Endophytic fungi could yield volatile organic compounds (VOCs—mainly hydrocarbons and other oxygenated compounds) and lignocellulolytic enzymes while growing on plant and agricultural residues. VOCs are being measured as promising alternatives to fossil as they look like fossil fuels and are known as mycodiesel (Strobel et al. 2008; Suryanarayanan et al. 2012; Wu et al. 2016). Endophytic fungi absorb nutrients from the plant as well as from the surrounding environment and hence secrete enzymes to catabolize complex organic matter (Thirunavukkarasu et al. 2015). Endophytes secreted enzymes can be exploited for biotechnological purposes by various industries (agriculture, pulp, pharmaceutical, paper, and industries) for example glutaminase-free l-asparaginase is used for the treatment of lymphoblastic leukemia (Nagarajan et al. 2014). Chitin-modifying enzymes secreted by fungal endophytes isolated from leaves of various Western Ghats trees can acetylate chitosan to diverse degrees (Rajulu et al. 2011). Compared with crustacean chitosans, the fungal enzymes are of low molecular weight, have higher polydispersity, and act on substrates with a lower degree of acetylation (Nwe et al. 2009). Therefore, screening for the diversity of such endophytic fungal enzymes is a novel but promising method to be utilized in food and environmental industries (Suryanarayanan et al. 2012) for example the fungal enzyme laccase can be exploited for the biosensor formation, biodegradation of dyes, phenols, and pesticides (Senthivelan et al. 2016). Pigment secreted by Penicillium aculeatum can be used in the soft drink because of its stability at neutral pH (Mapari et al. 2009) and pigment from Monascus purpureus is stable at higher pH (Huang et al. 2011). Natural colorants fungal endophytes have wide applications in cosmetics, foods, pharma, and textiles industries for example glucoamylase from Aspergillus niger and A. oryzae is exploited by the sugar and starch industry to harvest glucose (Devi et al. 2020). Due to increasing health consciousness, the use of fungal food like mycoproteins has risen globally as nutraceuticals, especially among vegetarian folks (Ghorai et al. 2009). Besides the food industry, traditional Chinese medicine has been using shiitake mushrooms for example Ganoderma lucidum is a promising anti-cancer agent, stress reducer, and potent immune system regulator (Devi et al. 2020).

A few examples of endophytic fungi involved in agriculture, bioremediation, biomedicine, pharmaceutical, biofuel, biocatalyst, and in the production of functional secondary metabolites, polysaccharides, are mentioned in Table 5.

Conclusion

Fungal endophytes oblige as “biosynthetic platforms” of significant secondary metabolites with huge biotechnological applications. However, partial knowledge of endophyte biology and reduced production of secondary metabolites in recurring sub-cultures of endophytic strains necessitates the requirement to adopt a comprehensive and systematic approach toward the exploitation of endophytes in various industries and research. In this direction, a forthcoming strategy is to use high-throughput technologies and the “genomic revolution” to develop strain improvement methods (Lautru et al. 2005). One such approach is the inclusion of metabolic pathways and genes in endophytes via genetic recombination between plant hosts and endophytes since endophytes mimic their host plant in the self-governing biosynthesis of secondary metabolites (Tiwari and Bae 2020). Scientific tools and approaches are intended to employ the co-integration of bioprocess techniques and genetic/metabolomics approaches to study the dynamics of plant–endophyte interactions and produce high yields of industrially important metabolites.

Acknowledgements

PT wants to express her gratitude to University Grants Commission (UGC), Govt. of India for providing MANF-UGC JRF fellowship (UGC-Ref. No.: 3536/CSIR-UGC NET JUNE 2019). PM is highly grateful to the University of North Bengal for providing the necessary facilities for writing this review article.

Author contributions

PT—writing, editing, formatting; LLK and AG—writing, formatting, preparation of figures and tables; PG—writing, editing; RC—writing; SR—editing, PM—conceptualization, drafting, editing, formatting, corrections.

Funding

This work was supported by the University of North Bengal with research project Ref. No. 2225/R-2021 granted to Dr. Piyush Mathur.

Availability of data and materials

Not applicable.

Declarations

Conflict of interest

All the authors declare that there is no conflict of interest.

Contributor Information

Prabha Toppo, Email: probha.gkt@gmail.com.

Lahasang Lamu Kagatay, Email: lahasanglamukagatay@gmail.com.

Ankita Gurung, Email: gurungankita39@gmail.com.

Priyanka Singla, Email: priyankabot@gmail.com, Email: priyankasingla@mccblr.edu.in.

Rakhi Chakraborty, Email: chakraborty.rakhi86@gmail.com.

Swarnendu Roy, Email: swarnendubotany@nbu.ac.in.

Piyush Mathur, Email: piyushmathur110@gmail.com, Email: piyushmathur316@nbu.ac.in.

References

  1. Abdalla MA, Aro AO, Gado D, Passari AK, Mishra VK, Singh BP, McGaw LJ. Isolation of endophytic fungi from South African plants, and screening for their anti-microbial and extracellular enzymatic activities and presence of type I polyketide synthases. S Afr J Bot. 2020 doi: 10.1016/j.sajb.2020.03.021. [DOI] [Google Scholar]
  2. Abdelwahab MF, Kurtán T, Mándi A, Müller WE, Fouad MA, Kamel MS, Liu Z, Ebrahim W, Daletos G, Proksch P. Induced secondary metabolites from the endophytic fungus Aspergillus versicolor through bacterial co-culture and OSMAC approaches. Tetrahedron Lett. 2018;59(27):2647–2652. doi: 10.1016/j.tetlet.2018.05.067. [DOI] [Google Scholar]
  3. Abro MA, Sun X, Li X, Jatoi GH, Guo LD. Biocontrol potential of fungal endophytes against Fusarium oxysporum f. sp. cucumerinum causing wilt in cucumber. Plant Pathol J. 2019;35(6):598. doi: 10.5423/PPJ.OA.05.2019.0129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Aghdam SA, Brown AMV. Deep learning approaches for natural product discovery from plant endophytic microbiomes. Environ Microbiome. 2021;16:6. doi: 10.1186/s40793-021-00375-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ahamed A, Ahring BK. Production of hydrocarbon compounds by endophytic fungi Gliocladium species grown on cellulose. Bioresour Technol. 2011;102(20):97189722. doi: 10.1016/j.biortech.2011.07.073. [DOI] [PubMed] [Google Scholar]
  6. Ahmad I, del Mar Jiménez-Gasco M, Luthe DS, Shakeel SN, Barbercheck ME. Endophytic Metarhizium robertsii promotes maize growth, suppresses insect growth, and alters plant defense gene expression. Biol Control. 2020;144:104167. doi: 10.1016/j.biocontrol.2019.104167. [DOI] [Google Scholar]
  7. Aimé S, Alabouvette C, Steinberg C, Olivain C. The endophytic strain Fusarium oxysporum Fo47: a good candidate for priming the defense responses in tomato roots. Mol Plant Microbe Interac. 2013;26(8):918–926. doi: 10.1094/mpmi-12-12-0290-r. [DOI] [PubMed] [Google Scholar]
  8. Aishwarya SA, Nagam N, Vijaya T, Netala RV. Screening and identification of heavy metal-tolerant endophytic fungi Lasiodiplodia theobromae from Boswellia ovalifoliolata an endemic plant of tirumala hills. Asian J Pharm Clin Res. 2017;10(3):488–491. doi: 10.22159/ajpcr.2017.v10i3.16697. [DOI] [Google Scholar]
  9. Akinola SA, Babalola OO. The importance of adverse soil microbiomes in the light of omics: implications for food safety. Plant Soil Environ. 2020;66(9):421–430. doi: 10.17221/118/2020-PSE. [DOI] [Google Scholar]
  10. Akiyama DY, Rocha MC, Costa JH, Malavazi I, Fill TP. The histone deacetylase clr3 regulates secondary metabolite production and growth under oxidative stress conditions in Penicillium brasilianum. BioRxiv. 2020 doi: 10.1101/2020.05.01.072108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Akone SH, Mándi A, Kurtán T, Hartmann R, Lin W, Daletos G, Proksch P. Inducing secondary metabolite production by the endophytic fungus Chaetomium sp. through fungal–bacterial co-culture and epigenetic modification. Tetrahedron. 2016;72(41):6340–6347. doi: 10.1016/j.tet.2016.08.022. [DOI] [Google Scholar]
  12. Al Abdallah Q, Ge W, Fortwendel JR. A simple and universal system for gene manipulation in Aspergillus fumigatus: in vitro-assembled Cas9-guide RNA ribonucleoproteins C. mSphere. 2017;2:e00446–e517. doi: 10.1128/msphere.00446-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Albright JC, Henke MT, Soukup AA, McClure RA, Thomson RJ, Keller NP, Kelleher NL. Large-scale metabolomics reveals a complex response of Aspergillus nidulans to epigenetic perturbation. ACS Chem Biol. 2015;10(6):1535–1541. doi: 10.1021/acschembio.5b00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Aldholmi M, Wilkinson B, Ganesan A. Epigenetic modulation of secondary metabolite profiles in Aspergillus calidoustus and Aspergillus westerdijkiae through histone deacetylase (HDAC) inhibition by vorinostat. J Antibiot. 2020;73(6):410–413. doi: 10.1038/s41429-020-0286-5. [DOI] [PubMed] [Google Scholar]
  15. Al-Rashdi FKH, Al-Sadi AM, Al-Riyamy BZ, Maharachchikumbura SS, Al-Sabahi JN, Velazhahan R. Endophytic fungi from the medicinal plant Aloe dhufarensis Lavranos exhibit antagonistic potential against phytopathogenic fungi. S Afr J Bot. 2020 doi: 10.1016/j.sajb.2020.05.022. [DOI] [Google Scholar]
  16. Avalos J, Limón MC. Fungal secondary metabolism. Encyclopedia. 2021;2(1):1–3. doi: 10.3390/encyclopedia2010001. [DOI] [Google Scholar]
  17. Ayob FW, Simarani K, Zainal Abidin N, Mohamad J. First report on a novel Nigrospora sphaerica isolated from Catharanthus roseus plant with anticarcinogenic properties. Microb Biotechnol. 2017;10(4):926–932. doi: 10.1111/1751-7915.12603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bai M, Gao CH, Liu K, Zhao LY, Tang ZZ, Liu YH. Two new benzophenones isolated from a mangrove-derived fungus Penicillium sp. J Antibiot. 2021;74(11):821–824. doi: 10.1038/s41429-021-00464-9. [DOI] [PubMed] [Google Scholar]
  19. Bajaj R, Huang Y, Gebrechristos S, Mikolajczyk B, Brown H, Prasad R, Varma A, 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):1–8. doi: 10.1038/s41598-018-26809-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bang S, Song JH, Lee D, Lee C, Kim S, Kang K, Lee JH, Shim SH. Neuroprotective secondary metabolite produced by an endophytic fungus, Neosartorya fischeri JS0553, isolated from Glehnia littoralis. J Agric Food Chem. 2019;67:1831–1838. doi: 10.1021/acs.jafc.8b05481. [DOI] [PubMed] [Google Scholar]
  21. Baral B, Akhgari A, Metsä-Ketelä M. Activation of microbial secondary metabolic pathways: avenues and challenges. Synth Syst Biotechnol. 2018;3(3):163–178. doi: 10.1016/j.synbio.2018.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Baron NC, Rigobelo EC. Endophytic fungi: a tool for plant growth promotion and sustainable agriculture. Mycology. 2022;13(1):39–55. doi: 10.1080/21501203.2021.1945699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Bedi A, Adholeya A, Deshmukh SK. Novel anti-cancer compounds from endophytic fungi. Curr Biotechnol. 2018;7(3):168–184. doi: 10.2174/2211550105666160622080354. [DOI] [Google Scholar]
  24. Behie SW, Bidochka MJ. Nutrient transfer in plant–fungal symbioses. Trends Plant Sci. 2014;19(11):734–740. doi: 10.1016/j.tplants.2014.06.007. [DOI] [PubMed] [Google Scholar]
  25. Behie SW, Bidochka MJ. Ubiquity of insect-derived nitrogen transfer to plants by endophytic insect-pathogenic fungi: an additional branch of the soil nitrogen cycle. Appl Environ Microbiol. 2014;80(5):1553–1560. doi: 10.1128/AEM.03338-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bezerra JD, Nascimento CC, Barbosa RD, da Silva DC, Svedese VM, Silva-Nogueira EB, Gomes BS, Paiva LM, Souza-Motta CM. Endophytic fungi from medicinal plant Bauhinia forficata: diversity and biotechnological potential. Braz J Microbiol. 2015;46:49–57. doi: 10.1590/S1517-838246120130657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Bhalkar BN, Patil SM, Govindwar SP. Camptothecine production by mixed fermentation of two endophytic fungi from Nothapodytes nimmoniana. Fungal Biol. 2016;6–7:873–883. doi: 10.1016/j.funbio.2016.04.003. [DOI] [PubMed] [Google Scholar]
  28. Bhambhani S, Kondhare KR, Giri AP. Diversity in chemical structures and biological properties of plant alkaloids. Molecules. 2021;26(11):3374. doi: 10.3390/molecules26113374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bibi S, Hussain A, Hamayun M, Rahman H, Iqbal A, Shah M, Ieshad M, Qasim M, Islam B. Bioremediation of hexavalent chromium by endophytic fungi; safe and improved production of Lactuca sativa L. Chemosphere. 2018;211:653–663. doi: 10.1016/j.chemosphere.2018.07.197. [DOI] [PubMed] [Google Scholar]
  30. Bilal L, Asaf S, Hamayun M, Gul H, Iqbal A, Ullah I, Lee IJ, Hussain A. Plant growth promoting endophytic fungi Aspergillus fumigatus TS1 and Fusarium proliferatum BRL1 produce gibberellins and regulates plant endogenous hormones. Symbiosis. 2018;76:117–127. doi: 10.1007/s13199-018-0545-4. [DOI] [Google Scholar]
  31. Bin C, Fangming Y, Zhi J. Mogroside V-producing endophytic fungi isolated from Siraitia grosvenorii. Planta Med. 2020 doi: 10.1055/a-1102-1168. [DOI] [PubMed] [Google Scholar]
  32. Bodede O, Kuali M, Prinsloo G, Moodley R, Govinden R. Anti-Pseudomonas aeruginosa activity of a C16-terpene dilactone isolated from the endophytic fungus Neofusicoccum luteum of Kigelia africana (Lam.) Sci Rep. 2022;12(1):1–1. doi: 10.1038/s41598-021-04747-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Brotman Y, Landau U, Cuadros-Inostroza A, Takayuki T, Fernie AR, Chet I, Viterbo A, Willmitzer L. Trichoderma-plant root colonization: escaping early plant defense responses and activation of the anti-oxidant machinery for saline stress tolerance. PLoS Pathog. 2013;9(3):e1003221. doi: 10.1371/journal.ppat.1003221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Budhiraja A, Nepali K, Sapra S, Gupta S, Kumar S, Dhar KL. Bioactive metabolites from an endophytic fungus of Aspergillus species isolated from seeds of Gloriosa superba Linn. J Med Chem. 2013;22(1):323–329. doi: 10.1007/s00044-012-0032-z. [DOI] [Google Scholar]
  35. Bushley KE, Raja R, Jaiswal P, Cumbie JS, Nonogaki M, Boyd AE, Owensby CA, Knaus BJ, Elser J, Miller D, Di Y. The genome of Tolypocladium inflatum: evolution, organization, and expression of the cyclosporin biosynthetic gene cluster. PLoS Genet. 2013;9(6):e1003496. doi: 10.1371/journal.pgen.1003496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Cacho RA, Tang Y, Chooi YH. Next-generation sequencing approach for connecting secondary metabolites to biosynthetic gene clusters in fungi. Front Microbiol. 2015;5:774. doi: 10.3389/fmicb.2014.00774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Camehl I, Drzewiecki C, Vadassery J, Shahollari B, Sherameti I, Forzani C, Munnik T, Hirt H, Oelmüller R. The OXI1 kinase pathway mediates Piriformospora indica-induced growth promotion in Arabidopsis. PLoS Pathog. 2011;7(5):e1002051. doi: 10.1371/journal.ppat.1002051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Cao D, Sun P, Bhowmick S, Wei Y, Guo B, Wei Y, Mur LA, Sun Z. Secondary metabolites of endophytic fungi isolated from Huperzia serrata. Fitoterapia. 2021;155:104970. doi: 10.1016/j.fitote.2021.104970. [DOI] [PubMed] [Google Scholar]
  39. Cary JW, Uka V, Han Z, Buyst D, Harris-Coward PY, Ehrlich KC, Wei Q, Bhatnagar D, Dowd PF, Martens SL, Calvo AM. An Aspergillus flavus secondary metabolic gene cluster containing a hybrid PKS–NRPS is necessary for synthesis of the 2-pyridones, leporins. Fungal Genet Biol. 2015;81:88–97. doi: 10.1016/j.fgb.2015.05.010. [DOI] [PubMed] [Google Scholar]
  40. Chakraborty P, Chakraborty A. Biosynthetic gene clusters in organism: the sole source of new drug discovery. J Mol Genet Med. 2021;15(2021):501. [Google Scholar]
  41. Chen XM, Sang XX, Li SH, Zhang SJ, Bai LH. Studies on a chlorogenic acid-producing endophytic fungi isolated from Eucommia ulmoides Oliver. J Ind Microbiol Biotechnol. 2010;37(5):447–454. doi: 10.1007/s10295-010-0690-0. [DOI] [PubMed] [Google Scholar]
  42. Chen Y, Mao W, Tao H, Zhu W, Qi X, Chen Y, Li H, Zhao C, Yang Y, Hou Y. Structural characterization and anti-oxidant properties of an exopolysaccharide produced by the mangrove endophytic fungus Aspergillus sp. Y16. Bioresour Technol. 2011;102(17):8179–8184. doi: 10.1016/j.biortech.2011.06.048. [DOI] [PubMed] [Google Scholar]
  43. Chen F, Tan N, Long W, Yang SK, She ZG, Lin YC. Enhancement of uranium (VI) biosorption by chemically modified marine-derived mangrove endophytic fungus Fusarium sp. #ZZF51. J Radioanal Nucl Chem. 2014;299(1):193–201. doi: 10.1007/s10967-013-2758-6. [DOI] [Google Scholar]
  44. Chen XW, Yang ZD, Sun JH, Song TT, Zhu BY, Zhao JW. Colletotrichine A, a new sesquiterpenoid from Colletotrichum gloeosporioides GT-7, a fungal endophyte of Uncaria rhynchophylla. Nat Prod Res. 2018;32:880–884. doi: 10.1080/14786419.2017.1365071. [DOI] [PubMed] [Google Scholar]
  45. Chen H, Qi Y, He X, Xu L, Zhang W, Lv X, Zhang H, Yang D, Zhu Y, Liang Z. Endophytic fungus Mucor circinelloides DF20 promote tanshinone biosynthesis and accumulation in Salvia miltiorrhiza root. Plant Sci. 2021;307:110898. doi: 10.1016/j.plantsci.2021.110898. [DOI] [PubMed] [Google Scholar]
  46. Chen H, Qi Y, He X, Xu L, Zhang W, Lv X, Zhang H, Yang D, Zhu Y, Liang Z. Endophytic fungus Mucor circinelloides DF20 promote tanshinone biosynthesis and accumulation in Salvia miltiorrhiza root. Plant Sci. 2021;307:110898. doi: 10.1016/j.plantsci.2021.110898. [DOI] [PubMed] [Google Scholar]
  47. Chen J, Li L, Tian P, Xiang W, Lu X, Huang R, Li L. Fungal endophytes from medicinal plant Bletilla striata (Thunb.) Reichb. F. promote the host plant growth and phenolic accumulation. South Afr J Bot. 2021;143:25–32. doi: 10.1016/j.sajb.2021.07.041. [DOI] [Google Scholar]
  48. Cheng JT, Cao F, Chen XA, Li YQ, Mao XM. Genomic and transcriptomic survey of an endophytic fungus Calcarisporium arbuscula NRRL 3705 and potential overview of its secondary metabolites. BMC Genom. 2020;21(1):1–3. doi: 10.1186/s12864-020-06813-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Chowdhary K, Kaushik N. Fungal endophyte diversity and bioactivity in the Indian medicinal plant Ocimum sanctum Linn. PLoS ONE. 2015;10:e0141444. doi: 10.1371/journal.pone.0141444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Cichewicz RH. Epigenome manipulation as a pathway to new natural product scaffolds and their congeners. Nat Prod Rep. 2010;27(1):11–22. doi: 10.1039/B920860G. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Courtial J, Helesbeux JJ, Oudart H, Aligon S, Bahut M, Hamon B, N’guyen G, Pigné S, Hussain AG, Pascouau C, Bataillé-Simoneau N. Characterization of NRPS and PKS genes involved in the biosynthesis of SMs in Alternaria dauci including the phytotoxic polyketide aldaulactone. Sci Rep. 2022;12(1):1–20. doi: 10.1038/s41598-022-11896-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Cretton S, Dorsaz S, Azzollini A, Favre-Godal Q, Marcourt L, Ebrahimi SN, Voinesco F, Michellod E, Sanglard D, Gindro K, Wolfender JL. Anti-fungal quinoline alkaloids from Waltheria indica. J Nat Prod. 2016;79(2):300–307. doi: 10.1021/acs.jnatprod.5b00896. [DOI] [PubMed] [Google Scholar]
  53. Cruz-Miranda OL, Folch-Mallol J, Martínez-Morales F, Gesto-Borroto R, Villarreal ML, Taketa AC. Identification of a Huperzine A-producing endophytic fungus from Phlegmariurus taxifolius. Mol Biol Rep. 2020;47:489–495. doi: 10.1007/s11033-019-05155-1. [DOI] [PubMed] [Google Scholar]
  54. Cui Y, Yi D, Bai X, Sun B, Zhao Y, Zhang Y. Ginkgolide B produced endophytic fungus (Fusarium oxysporum) isolated from Ginkgo biloba. Fitoterapia. 2012;83(5):913–920. doi: 10.1016/j.fitote.2012.04.009. [DOI] [PubMed] [Google Scholar]
  55. d’Errico G, Aloj V, Flematti GR, Sivasithamparam K, Worth CM, Lombardi N, Ritieni A, Marra R, Lorito M, Vinale F. Metabolites of a Drechslera sp. endophyte with potential as biocontrol and bioremediation agent. Nat Prod Res. 2021;35(22):4508–4516. doi: 10.1080/14786419.2020.1737058. [DOI] [PubMed] [Google Scholar]
  56. Dar RA, Saba I, Shahnawaz M, Qazi PH, Khan IA. Anti-microbial potential of fungal endophytes from selected high value medicinal plants of the Kashmir valley–India. J Psychopharmacol. 2017;6:307–310. [Google Scholar]
  57. Das AK. Anti-cancer effect of antimalarial artemisinin compounds. Ann Med Health Sci Res. 2015;5(2):93–102. doi: 10.4103/2141-9248.153609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Das A, Kamal S, Shakil NA, Sherameti I, Oelmüller R, Dua M, Tuteja N, Johri AK, 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):103–112. doi: 10.4161/psb.7.1.18472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. de Siqueira KA, Brissow ER, Santos JL, White JF, Santos FR, de Almeida EG, Soares MA. Endophytism and bioactivity of endophytic fungi isolated from Combretum lanceolatum Pohl ex Eichler. Symbiosis. 2017;71(3):211–222. doi: 10.1007/s13199-016-0427-6. [DOI] [Google Scholar]
  60. Deka D, Jha D. Anti-microbial activity of endophytic fungi from leaves and barks of Litsea cubeba Pers., a traditionally important medicinal plant of North East India. Jordan J Biol Sci. 2018;11:73–79. [Google Scholar]
  61. Deng Z, Cao L. Fungal endophytes and their interactions with plants in phytoremediation: a review. Chemosphere. 2017;168:1100–1106. doi: 10.1016/j.chemosphere.2016.10.097. [DOI] [PubMed] [Google Scholar]
  62. Deng Z, Zhang R, Shi Y, Hu L, Tan H, Cao L. Characterization of Cd-, Pb-, Zn-resistant endophytic Lasiodiplodia sp. MXSF31 from metal accumulating Portulaca oleracea and its potential in promoting the growth of rape in metal-contaminated soils. Environ Sci Pollut Res Int. 2014;21(3):2346–2357. doi: 10.1007/s11356-013-2163-2. [DOI] [PubMed] [Google Scholar]
  63. Deshmukh SK, Mishra PD, Kulkarni-Almeida A, Verekar S, Sahoo MR, Periyasamy G, Goswami H, Khanna A, Balakrishnan A, Vishwakarma R. Anti-inflammatory and anti-cancer activity of ergoflavin isolated from an endophytic fungus. Chem Biodivers. 2009;6(5):784–789. doi: 10.1002/cbdv.200800103. [DOI] [PubMed] [Google Scholar]
  64. Devari S, Jaglan S, Kumar M, Deshidi R, Guru S, Bhushan S, Kushwaha M, Gupta AP, Gandhi SG, Sharma JP, Taneja SC. Capsaicin production by Alternaria alternata, an endophytic fungus from Capsicum annum; LC–ESI–MS/MS analysis. Phytochemistry. 2014;98:183–189. doi: 10.1016/j.phytochem.2013.12.001. [DOI] [PubMed] [Google Scholar]
  65. Devi P, Rodrigues C, Naik CG, D’souza L. Isolation and characterization of anti-bacterial compound from a mangrove-endophytic fungus, Penicillium chrysogenum MTCC 5108. Indian J Microbiol. 2012;52(4):617–623. doi: 10.1007/s12088-012-0277-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Devi R, Kaur T, Guleria G, Rana KL, Kour D, Yadav N, Yadav AN, Saxena AK. Fungal secondary metabolites and their biotechnological applications for human health. In: Rastegari AA, Yadav AN, Yadav N, editors. New and future developments in microbial biotechnology and bioengineering. Amsterdam: Elsevier; 2020. pp. 147–161. [Google Scholar]
  67. Devi R, Abdulhaq A, Verma R, Sharma K, Kumar D, Kumar A, Tapwal A, Yadav R, Mohan S. Improvement in the phytochemical content and biological properties of Stevia rebaudiana (Bertoni) bertoni plant using endophytic fungi Fusarium fujikuroi. Plants (basel) 2023;12(5):1151. doi: 10.3390/plants12051151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Devi R, Verma R, Dhalaria R, Kumar A, Kumar D, Puri S, Thakur M, Chauhan S, Chauhan PP, Nepovimova E, Kuca K. A systematic review on endophytic fungi and its role in the commercial applications. Planta. 2023 doi: 10.1007/s00425-023-04087-2. [DOI] [PubMed] [Google Scholar]
  69. 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):362–369. doi: 10.1016/j.bjm.2017.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Ding Z, Tao T, Wang L, Zhao Y, Huang H, Zhang D, Liu M, Wang Z, Han J. Bioprospecting of novel and bioactive metabolites from endophytic fungi isolated from rubber tree Ficus elastica leaves. J Microbial Biotechnol. 2019;29:731–738. doi: 10.4014/jmb.1901.01015. [DOI] [PubMed] [Google Scholar]
  71. Ding Z, Zhou H, Wang X, Huang H, Wang H, Zhang R, Wang Z, Han J. Deletion of the histone deacetylase HdaA in endophytic fungus Penicillium chrysogenum Fes1701 induces the complex response of multiple bioactive secondary metabolite production and relevant gene cluster expression. Molecules. 2020;25:3657. doi: 10.3390/molecules25163657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Dissanayake AJ, Purahong W, Wubet T, Hyde KD, Zhang W, Xu H, Zhang G, Fu G, Liu M, Xing Q, Li X, Yan J. Direct comparison of culture-dependent and culture-independent molecular approaches reveal the diversity of fungal endophytic communities in stems of grapevine (Vitis vinifera) Fung Div. 2018;90:85–107. doi: 10.1007/s13225-018-0399-3. [DOI] [Google Scholar]
  73. Dos Santos IR, Abdel-Azeem AM, Mohesien MT, Piekutowska M, Sheir DH, da Silva LL, da Silva CC, Carvalho DD, Bezerra JD, Saad HA, Borges LL. Insights into the bioprospecting of the endophytic fungi of the medicinal plant Palicourea rigida Kunth (Rubiaceae): detailed biological activities. J Fungi. 2021;7(9):689. doi: 10.3390/jof7090689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Ehsan T, Reza RN, Das A, Ahmed O, Baten AA, Ferdous AS, Islam MR, Khan H. Genome and secretome analysis of jute endophyte Grammothele lineata strain SDL-CO-2015-1: insights into its lignocellulolytic structure and secondary metabolite profile. Genomics. 2020;112(4):2794–2803. doi: 10.1016/j.ygeno.2020.03.017. [DOI] [PubMed] [Google Scholar]
  75. El Hajj AC, Zetina-Serrano C, Tahtah N, Khoury AE, Atoui A, Oswald IP, Puel O, Lorber S. Regulation of secondary metabolism in the penicillium genus. Int J Mol Sci. 2020;21(24):9462. doi: 10.3390/ijms21249462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. El-Bialy HA, El-Bastawisy HS. Elicitors stimulate paclitaxel production by endophytic fungi isolated from ecologically altered Taxus baccata. J Radiat Res Appl Sci. 2020;13:79–87. doi: 10.1080/16878507.2019.1702244. [DOI] [Google Scholar]
  77. Elfita E, Widjajanti H, Setiawan A, Kurniawati A. Anti-bacterial activity of endophytic fungi isolated from the stem bark of jambu mawar (Syzygium jambos) Biodiversitas J Biol Divers. 2022 doi: 10.13057/biodiv/d230156. [DOI] [Google Scholar]
  78. Elkhouly HI, Hamed AA, El Hosainy AM, Ghareeb MA, Sidkey NM. Bioactive secondary metabolite from endophytic Aspergillus Tubenginses ASH4 isolated from Hyoscyamus muticus: anti-microbial, antibiofilm, anti-oxidant and anti-cancer activity. Pharmacogn J. 2021 doi: 10.5530/pj.2021.13.55. [DOI] [Google Scholar]
  79. Elkhouly HI, Sidkey NM, Ghareeb MA, El Hosainy AM, Hamed AA. Bioactive secondary metabolites from endophytic Aspergillus terreus AH1 isolated from Ipomoea carnea growing in Egypt. Egypt J Chem. 2021;64(12):7511–7520. doi: 10.21608/ejchem.2021.85908.4161. [DOI] [Google Scholar]
  80. El-Sayed ES, Zaki AG, Ahmed AS, Ismaiel AA. Production of the anti-cancer drug taxol by the endophytic fungus Epicoccum nigrum TXB502: enhanced production by gamma irradiation mutagenesis and immobilization technique. Appl Microbiol Biotechnol. 2020;104(16):6991–7003. doi: 10.1007/s00253-020-10712-x. [DOI] [PubMed] [Google Scholar]
  81. Erb M, Kliebenstein DJ. Plant secondary metabolites as defenses, regulators, and primary metabolites: the blurred functional trichotomy. Plant Physiol. 2020;184(1):39–52. doi: 10.1104/PP.20.00433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Fadiji AE, Babalola OO. Metagenomics methods for the study of plant-associated microbial communities: a review. J Microbiological Methods. 2020;170:105860. doi: 10.1016/j.mimet.2020.105860. [DOI] [PubMed] [Google Scholar]
  83. Fouda AH, Hassan SE, Eid AM, Ewais EE. Biotechnological applications of fungal endophytes associated with medicinal plant Asclepias sinaica (Bioss.) Ann Agric Sci. 2015;60(1):95–104. doi: 10.1016/j.aoas.2015.04.001. [DOI] [Google Scholar]
  84. Freitas F, Alves VD, Reis MA. Advances in bacterial exopolysaccharides: from production to biotechnological applications. Trends Biotechnol. 2011;29(8):388–398. doi: 10.1016/j.tibtech.2011.03.008. [DOI] [PubMed] [Google Scholar]
  85. Galle M, Crespo R, Rodenak Kladniew B, Montero Villegas S, Polo M, de Bravo MG. Suppression by geraniol of the growth of A549 human lung adenocarcinoma cells and inhibition of the mevalonate pathway in culture and in vivo: potential use in cancer chemotherapy. Nutr Cancer. 2014;66(5):888–895. doi: 10.1080/01635581. [DOI] [PubMed] [Google Scholar]
  86. Gao SS, Li XM, Williams K, Proksch P, Ji NY, Wang BG. Rhizovarins A-F, indole-diterpenes from the mangrove-derived endophytic fungus Mucor irregularis QEN-189. J Nat Prod. 2016;79:2066–2074. doi: 10.1021/acs.jnatprod.6b00403. [DOI] [PubMed] [Google Scholar]
  87. Gao T, Qin D, Zuo S, Peng Y, Xu J, Yu B, Song H, Dong J. Decolorization and detoxification of triphenylmethane dyes by isolated endophytic fungus, Bjerkandera adusta SWUSI4 under non-nutritive conditions. Bioresour Bioprocess. 2020;7(1):1–2. doi: 10.1186/s40643-020-00340-8. [DOI] [Google Scholar]
  88. Gautam VS, Singh A, Kumari P, Nishad JH, Kumar J, Yadav M, Bharti R, Prajapati P, Kharwar RN. Phenolic and flavonoid contents and anti-oxidant activity of an endophytic fungus Nigrospora sphaerica (EHL2), inhabiting the medicinal plant Euphorbia hirta (dudhi) L. Arch Microbiol. 2022;204(2):1–3. doi: 10.1007/s00203-021-02650-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Ghaffari MR, Mirzaei M, Ghabooli M, Khatabi B, Wu Y, Zabet-Moghaddam M, Mohammadi-Nejad G, Haynes PA, Hajirezaei MR, Sepehri M, Salekdeh GH. Root endophytic fungus Piriformospora indica improves drought stress adaptation in barley by metabolic and proteomic reprogramming. Environ Exp Bot. 2019;157:197–210. doi: 10.1016/j.envexpbot.2018.10.002. [DOI] [Google Scholar]
  90. Ghorai S, Banik SP, Verma D, Chowdhury S, Mukherjee S, Khowala S. Fungal biotechnology in food and feed processing. Food Res Int. 2009;42(5–6):577–587. doi: 10.1016/j.foodres.2009.02.019. [DOI] [Google Scholar]
  91. Gouda S, Das G, Sen SK, Shin HS, Patra JK. Endophytes: a treasure house of bioactive compounds of medicinal importance. Front Microbiol. 2016;7:1538. doi: 10.3389/fmicb.2016.01538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Griffin MA, Spakowicz DJ, Gianoulis TA, Strobel SA. Volatile organic compound production by organisms in the genus Ascocoryne and a re-evaluation of myco-diesel production by NRRL 50072. Microbiology. 2010;156(12):3814–3829. doi: 10.1099/mic.0.041327-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Grijseels S, Pohl C, Nielsen JC, Wasil Z, Nygård Y, Nielsen J, Frisvad JC, Nielsen KF, Workman M, Larsen TO, Driessen AJ. Identification of the decumbenone biosynthetic gene cluster in Penicillium decumbens and the importance for production of calbistrin. Fungal Biol Biotechnol. 2018;5(1):1–7. doi: 10.1186/s40694-018-0063-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Gupta N, Sabat J, Parida R, Kerkatta D. Solubilization of tricalcium phosphate and rock phosphate by microbes isolated from chromite, iron and manganese mines. Acta Bot Croat. 2007;66(2):197–204. [Google Scholar]
  95. Gupta S, Chaturvedi P (2019) Enhancing secondary metabolite production in medicinal plants using endophytic elicitors: a case study of Centella asiatica (Apiaceae) and asiaticoside. In: Hodkinson TR, Saunders MJ, Murphy BR, Dublin TC, Doohan FM (eds) Endophytes for a growing world, vol 21, pp 310–323
  96. Hamayun M, Afzal Khan S, Ahmad N, Tang DS, Kang SM, Na CI, Sohn EY, Hwang YH, Shin DH, Lee BH, Kim JG. Cladosporium sphaerospermum as a new plant growth-promoting endophyte from the roots of Glycine max (L.) Merr. World J Microbiol Biotechnol. 2009;25(4):627–632. doi: 10.1007/s11274-009-9982-9. [DOI] [Google Scholar]
  97. Han D, Wang K, Long F, Zhang W, Yao X, Chen S. Effects of endophytic fungi on the secondary metabolites of Hordeum bogdanii under alkaline stress. AMB Express. 2022;12(1):1–9. doi: 10.1186/s13568-022-01414-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. 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. doi: 10.1128/MMBR.00050-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Harnpicharnchai P, Champreda V, Sornlake W, Eurwilaichitr L. A thermotolerant β-glucosidase isolated from an endophytic fungus, Periconia sp., with a possible use for biomass conversion to sugars. Protein Expr Purif. 2009;67(2):61–69. doi: 10.1016/j.pep.2008.05.022. [DOI] [PubMed] [Google Scholar]
  100. Harwoko H, Daletos G, Stuhldreier F, Lee J, Wesselborg S, Feldbrügge M, Muller WE, Kalscheuer R, Ancheeva E, Proksch P. Dithiodiketopiperazine derivatives from endophytic fungi Trichoderma harzianum and Epicoccum nigrum. Nat Prod Res. 2019 doi: 10.1080/14786419.2019.1627348. [DOI] [PubMed] [Google Scholar]
  101. Hassan TU, Bano A, Naz I. Alleviation of heavy metals toxicity by the application of plant growth promoting rhizobacteria and effects on wheat grown in saline sodic field. Int J Phytoremediation. 2017;19(6):522–529. doi: 10.1080/15226514.2016.1267696. [DOI] [PubMed] [Google Scholar]
  102. Hassane AM, Taha TM, Awad MF, Mohamed H, Melebari M. Radical scavenging potency, HPLC profiling and phylogenetic analysis of endophytic fungi isolated from selected medicinal plants of Saudi Arabia. Electron J Biotechnol. 2022 doi: 10.1016/j.ejbt.2022.05.001. [DOI] [Google Scholar]
  103. Hiranrat W, Hiranrat A, Supaphon P. Anti-microbial secondary metabolites from endophytic fungus Fusarium sp. from Eichhornia crassipes Linn. Asean J Sci Technol Rep. 2021 doi: 10.55164/ajstr.v24i3.244115. [DOI] [Google Scholar]
  104. Huang CH, Pan JH, Chen B, Yu M, Huang HB, Zhu X, Lu YJ, She ZG, Lin YC. Three bianthraquinone derivatives from the mangrove endophytic fungus Alternaria sp. ZJ9-6B from the South China Sea. Mar Drugs. 2011;9(5):832–843. doi: 10.3390/md9050832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Huang JX, Zhang J, Zhang XR, Zhang K, Zhang X, He XR. Mucor fragilis as a novel source of the key pharmaceutical agents podophyllotoxin and kaempferol. Pharm Biol. 2014;52(10):1237–1243. doi: 10.3109/13880209.2014.885061. [DOI] [PubMed] [Google Scholar]
  106. Huang Q, An H, Song H, Mao H, Shen W, Dong J. Diversity and biotransformative potential of endophytic fungi associated with the medicinal plant Kadsura angustifolia. Res Microbiol. 2015;166:45–55. doi: 10.1016/j.resmic.2014.12.004. [DOI] [PubMed] [Google Scholar]
  107. Huang S, Chen H, Li W, Zhu X, Ding W, Li C. Bioactive chaetoglobosins from the mangrove endophytic fungus Penicillium chrysogenum. Mar Drugs. 2016;14:172. doi: 10.3390/md14100172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Huang X, Zhang W, Tang S, Wei S, Lu X. Collaborative biosynthesis of a class of bioactive azaphilones by two separate gene clusters containing four PKS/NRPSs with transcriptional crosstalk in fungi. Angew Chem. 2020;132(11):4379–4383. doi: 10.1002/ange.201915514. [DOI] [PubMed] [Google Scholar]
  109. Huang X, Zeng Z, Chen Z, Yang Y, Pang J, Qian Y, Xiang T. Whole-Genome Sequence Resource of Fusarium oxysporum Strain TH15, a Plant Growth Promoting Endophytic Fungus Isolated from Tetrastigma hemsleyanum. PhytoFrontiers™ PHYTOFR-12. 2022 doi: 10.1094/PHYTOFR-12-21-0086-A. [DOI] [Google Scholar]
  110. Hussain T, Javed M, Shaikh S, Tabasum B, Hussain K, Ansari MS, Khan A, Khan AA. Biotechnological approaches to enhance plant secondary metabolites. New York: CRC Press; 2021. Enhancement of plant secondary metabolites using fungal endophytes; pp. 61–70. [Google Scholar]
  111. Hussain T, Javed M, Shaikh S, Tabasum B, Hussain K, Ansari MS, Khan A, Khan AA. Enhancement of plant secondary metabolites using fungal endophytes. In: Shahnawaz M, editor. Biotechnological approaches to enhance plant secondary metabolites. New York: CRC Press; 2021. pp. 61–70. [Google Scholar]
  112. Hussein RA, El-Anssary AA. Plants secondary metabolites: the key drivers of the pharmacological actions of medicinal plants. Herb Med. 2019 doi: 10.5772/intechopen.76139. [DOI] [Google Scholar]
  113. Igiehon NO, Babalola OO. Biofertilizers and sustainable agriculture: exploring arbuscular mycorrhizal fungi. Appl Microbiol Biotechnol. 2017;101:4871–4881. doi: 10.1007/s00253-017-8344-z. [DOI] [PubMed] [Google Scholar]
  114. Isah T, Umar S, Mujib A, Sharma MP, Rajasekharan PE, Zafar N, Frukh A. Secondary metabolism of pharmaceuticals in the plant in vitro cultures: strategies, approaches, and limitations to achieving higher yield. Plant Cell Tissue Organ Cult. 2018;132:239–265. doi: 10.1007/s11240-017-1332-2. [DOI] [Google Scholar]
  115. Iven T, König S, Singh S, Braus-Stromeyer SA, Bischoff M, Tietze LF, Braus GH, Lipka V, Feussner I, Dröge-Laser W. Transcriptional activation and production of tryptophan-derived secondary metabolites in Arabidopsis roots contributes to the defense against the fungal vascular pathogen Verticillium longisporum. Mol Plant. 2012;5:1389–1402. doi: 10.1093/mp/sss044. [DOI] [PubMed] [Google Scholar]
  116. Jayaram H, Marigowda V, Thara Saraswathi KJ. Secondary metabolite production and terpenoid biosynthesis in endophytic fungi Cladosporium cladosporioides Isolated from Wild Cymbopogon martinii (Roxb.) Wats. Microbiol Res. 2021;12(4):812–828. doi: 10.3390/microbiolres12040059. [DOI] [Google Scholar]
  117. Jayasekara C, Poonsawad A, Watchaputi K, Wattanachaisaereekul S, Soontorngun N. Media optimization of anti-microbial activity production and beta-glucan content of endophytic fungi Xylaria sp. BCC 1067. Biotechnol Rep. 2022 doi: 10.1016/j.btre.2022.e00742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Jiang Y, Wu J, Kawagishi H, Jiang C, Zhou Q, Tong Z, Tong Y, Wang P. Study on secondary metabolites of endophytic fungus, Aspergillus fumigatus, from Crocus sativus L. guided by UHPLC-HRMS/MS-based molecular network. Int J Anal Chem. 2022 doi: 10.1155/2022/7067665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Jin Z, Gao L, Zhang L, Liu T, Yu F, Zhang Z, Guo Q, Wang B. Anti-microbial activity of saponins produced by two novel endophytic fungi from Panax notoginseng. Nat Prod Res. 2017;31:2700–2703. doi: 10.1080/14786419.2017.1292265. [DOI] [PubMed] [Google Scholar]
  120. Johnson LJ, de Bonth A, Briggs LR, Caradus JR, Finch SC, Fleetwood DJ, Fletcher LR, Hume DE, Johnson RD, Popay AJ, Tapper BA. The exploitation of epichloae endophytes for agricultural benefit. Fungal Diver. 2013;60(1):171–188. doi: 10.1007/s13225-013-0239-4. [DOI] [Google Scholar]
  121. Kalimuthu AK, Parasuraman P, Sivakumar P, Murugesan S, Arunachalam S, Pandian SR, Ravishankar V, Ammunje DN, Sampath M, Panneerselvam T, Kunjiappan S. In silico, in vitro screening of anti-oxidant and anti-cancer potentials of bioactive secondary metabolites from an endophytic fungus (Curvularia sp.) from Phyllanthus niruri L. Environ Sci Pollut Res. 2022 doi: 10.1007/s11356-022-19249-0. [DOI] [PubMed] [Google Scholar]
  122. Kamel NM, Abdel-Motaal FF, El-Zayat SA. Endophytic fungi from the medicinal herb Euphorbia geniculata as a potential source for bioactive metabolites. Arch Microbiol. 2020;202:247–255. doi: 10.1007/s00203-019-01740-x. [DOI] [PubMed] [Google Scholar]
  123. Kang X, Liu C, Shen P, Hu L, Lin R, Ling J, Xiong X, Xie B, Liu D. Genomic characterization provides new insights into the biosynthesis of the secondary metabolite huperzine a in the endophyte Colletotrichum gloeosporioides Cg01. Front Microbiol. 2019;9:3237. doi: 10.3389/fmicb.2018.03237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Karimi R, Amini H, Ghabooli M. Root endophytic fungus Piriformospora indica and zinc attenuate cold stress in grapevine by influencing leaf phytochemicals and minerals content. Sci Hortic. 2022;293:110665. doi: 10.1016/j.scienta.2021.110665. [DOI] [Google Scholar]
  125. Katoch M, Pull S. Endophytic fungi associated with Monarda citriodora, an aromatic and medicinal plant and their biocontrol potential. Pharm Biol. 2017;55:1528–1535. doi: 10.1080/13880209.2017.1309054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Kaul S, Ahmed M, Zargar K, Sharma P, Dhar MK. Prospecting endophytic fungal assemblage of Digitalis lanata Ehrh. (foxglove) as a novel source of digoxin: a cardiac glycoside. 3 Biotech. 2013;3(4):335–340. doi: 10.1007/s13205-012-0106-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Kaushik NK, Murali TS, Sahal D, Suryanarayanan TS. A search for antiplasmodial metabolites among fungal endophytes of terrestrial and marine plants of southern India. Acta Parasitol. 2014;59(4):745–757. doi: 10.2478/s11686-014-0307-2. [DOI] [PubMed] [Google Scholar]
  128. Keller NP. Fungal secondary metabolism: regulation, function and drug discovery. Nat Rev Microbiol. 2019;17(3):167–180. doi: 10.1038/s41579-018-0121-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Khalil AM, Hassan SE, Alsharif SM, Eid AM, Ewais EE, Azab E, Gobouri AA, Elkelish A, Fouda A. Isolation and characterization of fungal endophytes isolated from medicinal plant Ephedra pachyclada as plant growth-promoting. Biomolecules. 2021;11(2):140. doi: 10.3390/biom11020140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Khan AR, Ullah I, Waqas M, Shahzad R, Hong SJ, Park GS, Jung BK, Lee IJ, Shin JH. Plant growth-promoting potential of endophytic fungi isolated from Solanum nigrum leaves. World J Microbiol Biotechnol. 2015;31(9):1461–1466. doi: 10.1007/s11274-015-1888-0. [DOI] [PubMed] [Google Scholar]
  131. Khan AR, Ullah I, Waqas M, Park GS, Khan AL, Hong SJ, Ullah R, Jung BK, Park CE, Ur-Rehman S. Host plant growth promotion and cadmium detoxification in Solanum nigrum, mediated by endophytic fungi. Ecotoxicol Environ Saf. 2017;136(2017):180–188. doi: 10.1016/j.ecoenv.2016.03.014. [DOI] [PubMed] [Google Scholar]
  132. Khan MS, Gao J, Munir I, Zhang M, Liu Y, Xue J, Zhang X. Characterization of endophytic fungi, Acremonium sp., from Lilium davidii and analysis of its anti-fungal and plant growth-promoting effects. BioMed Res Int. 2021 doi: 10.1155/2021/9930210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Kharwar RN, Verma VC, Kumar A, Gond SK, Harper JK, Hess WM, Lobkovosky E, Ma C, Ren Y, Strobel GA. Javanicin, an anti-bacterial naphthaquinone from an endophytic fungus of neem Chloridium sp. Curr Microbiol. 2009;58(3):233–238. doi: 10.1007/s00284-008-9313-7. [DOI] [PubMed] [Google Scholar]
  134. Khattak SU, Ur Rehman H, Khurshid A, Ijaz B. Anti-bacterial and anti-fungal potential of endophytic fungi isolated from different tissues of selected medicinal plants. Pure Appl Biol. 2020;9:989–998. doi: 10.19045/bspab.2020.90104. [DOI] [Google Scholar]
  135. Khiralla A, Mohamed I, Thomas J, Mignard B, Spina R, Yagi S, Laurain-Mattar D. A pilot study of anti-oxidant potential of endophytic fungi from some Sudanese medicinal plants. Asian Pac J Trop Med. 2015;8(9):701–704. doi: 10.1016/j.apjtm.2015.07.032. [DOI] [PubMed] [Google Scholar]
  136. Khiralla A, Mohamed IE, Tzanova T, Schohn H, Slezack-Deschaumes S, Hehn A, André P, Carre G, Spina R, Lobstein A, Yagi S. Endophytic fungi associated with Sudanese medicinal plants show cytotoxic and antibiotic potential. FEMS Microbiol Lett. 2016 doi: 10.1093/femsle/fnw089. [DOI] [PubMed] [Google Scholar]
  137. Khuseib Hamed Al-Rashdi F, Al-Sadi AM, Al-Riyamy BZ, Maharachchikumbura SN, Khalfan Al-Ruqaishi H, Velazhahan R. Alternaria alternata and Neocosmospora sp. from the medicinal plant Euphorbia larica exhibit antagonistic activity against Fusarium sp., a plant pathogenic fungus. All Life. 2020;13:223–232. doi: 10.1080/26895293.2020.1759702. [DOI] [Google Scholar]
  138. Kiers ET, Duhamel M, Beesetty Y, Mensah JA, Franken O, Verbruggen E, Fellbaum CR, Kowalchuk GA, Hart MM, Bago A, Palmer TM, West SA, Vandenkoornhuyse P, Jansa J, Bücking H. Reciprocal rewards stabilize cooperation in the mycorrhizal symbiosis. Science. 2011;333(6044):880–882. doi: 10.1126/science.1208473. [DOI] [PubMed] [Google Scholar]
  139. Kim JH, Yi YS, Kim MY, Cho JY. Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J Ginseng Res. 2017;41(4):435–443. doi: 10.1016/j.jgr.2016.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Kjærbølling I, Vesth TC, Frisvad JC, Nybo JL, Theobald S, Kuo A, Bowyer P, Matsuda Y, Mondo S, Lyhne EK, Kogle ME. Linking secondary metabolites to gene clusters through genome sequencing of six diverse Aspergillus species. Proc Natl Acad Sci. 2018;115(4):E753–E761. doi: 10.1073/pnas.1715954115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Kjærbølling I, Mortensen UH, Vesth T, Andersen MR. Strategies to establish the link between biosynthetic gene clusters and secondary metabolites. Fungal Genet Biol. 2019;130:107–121. doi: 10.1016/j.fgb.2019.06.001. [DOI] [PubMed] [Google Scholar]
  142. Kong YR, Tay KC, Su YX, Wong CK, Tan WN, Khaw KY. Potential of naturally derived alkaloids as multi-targeted therapeutic agents for neurodegenerative diseases. Molecules. 2021;26(3):728. doi: 10.3390/molecules26030728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Kongprapan T, Rukachaisirikul V, Saithong S, Phongpaichit S, Poonsuwan W, Sakayaroj J. Cytotoxic cytochalasins from the endophytic fungus Eutypella scoparia PSU-H267. Phytochem Lett. 2015;13:171–176. doi: 10.1016/j.phytol.2015.06.010. [DOI] [Google Scholar]
  144. Krell V, Unger S, Jakobs-Schoenwandt D, Patel AV. Endophytic Metarhizium brunneum mitigates nutrient deficits in potato and improves plant productivity and vitality. Fungal Ecol. 2018;34:43–49. doi: 10.1016/j.funeco.2018.04.002. [DOI] [Google Scholar]
  145. Kumaresan S, Karthi V, Senthilkumar V, Balakumar BS, Stephen A. Biochemical constituents and anti-oxidant potential of endophytic fungi isolated from the leaves of Azadirachta indica A. Juss (Neem) from Chennai, India. J Acad Ind Res. 2015;3:355–361. [Google Scholar]
  146. Kumari A, Singh D, Kumar S. Biotechnological interventions for harnessing podophyllotoxin from plant and fungal species: current status, challenges, and opportunities for its commercialization. Crit Rev Biotech. 2016;37(6):739–753. doi: 10.1080/07388551.2016.1228597. [DOI] [PubMed] [Google Scholar]
  147. Kusari S, Zühlke S, Spiteller M. Effect of artificial reconstitution of the interaction between the plant Camptotheca acuminata and the fungal endophyte Fusarium solani on camptothecin biosynthesis. J Nat Prod. 2011;74(4):764–775. doi: 10.1021/np1008398. [DOI] [PubMed] [Google Scholar]
  148. Kusari S, Verma VC, Lamshoeft M, Spiteller M. An endophytic fungus from Azadirachta indica A. Juss. that produces azadirachtin. World J Microbiol Biotechnol. 2012;28(3):1287–1294. doi: 10.1007/s11274-011-0876-2. [DOI] [PubMed] [Google Scholar]
  149. Kwon MJ, Steiniger C, Cairns TC, Wisecaver JH, Lind AL, Pohl C, Regner C, Rokas A, Meyer V. Beyond the biosynthetic gene cluster paradigm: genome-wide coexpression networks connect clustered and unclustered transcription factors to secondary metabolic pathways. Microbiol Spectr. 2021;9(2):e00898–e1821. doi: 10.1128/Spectrum.00898-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Lautru S, Deeth RJ, Bailey LM, Challis GL. Discovery of a new peptide natural product by Streptomyces coelicolor genome mining. Nat Chem Biol. 2005;1:265–269. doi: 10.1038/nchembio731. [DOI] [PubMed] [Google Scholar]
  151. Le TTM, Hoang ATH, Nguyen NP, Le TTB, Trinh HTT, Vo TTB, Trinh HT, Vo TT, Van Quyen D. A novel huperzine A-producing endophytic fungus Fusarium sp. Rsp5. 2 isolated from Huperzia serrate. Biotechnol Lett. 2020;42:987–995. doi: 10.1007/s10529-020-02836-x. [DOI] [PubMed] [Google Scholar]
  152. Lebar MD, Cary JW, Majumdar R, Carter-Wientjes CH, Mack BM, Wei Q, Uka V, De Saeger S, Di Mavungu JD. Identification and functional analysis of the aspergillic acid gene cluster in Aspergillus flavus. Fungal Genet Biol. 2018;116:14–23. doi: 10.1016/j.fgb.2018.04.009. [DOI] [PubMed] [Google Scholar]
  153. 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(2):233–229. doi: 10.1007/s00253-009-1856-4. [DOI] [PubMed] [Google Scholar]
  154. Li P, Mou Y, Shan T, Xu J, Li Y, Lu S, Zhou L. Effects of polysaccharide elicitors from endophytic Fusarium oxysporium Dzf17 on growth and diosgenin production in cell suspension culture of Dioscorea zingiberensis. Molecules. 2011;16(11):9003–9016. doi: 10.3390/molecules16119003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Li P, Lu S, Shan T, Mou Y, Li Y, Sun W, Zhou L. Extraction optimization of water-extracted mycelial polysaccharide from endophytic fungus Fusarium oxysporum Dzf17 by response surface methodology. Int J Mol Sci. 2012;13(5):5441–5453. doi: 10.3390/ijms13055441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Li P, Xu L, Mou Y, Shan T, Mao Z, Lu S, Peng Y, Zhou L. Medium optimization for exopolysaccharide production in liquid culture of endophytic fungus Berkleasmium sp. Dzf12. Int J Mol Sci. 2012;13(9):11411–11426. doi: 10.3390/ijms130911411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Li YL, Yi JL, Cai J, Zhou XM, Chen L, Zhuo X, Lai XY. Two new bioactive secondary metabolites from the endophytic fungus Talaromyces assiutensis JTY2. Nat Prod Res. 2021;30:1–6. doi: 10.1080/14786419.2021.1881961. [DOI] [PubMed] [Google Scholar]
  158. Liang Z, Zhang T, Zhang X, Zhang J, Zhao C. An alkaloid and a steroid from the endophytic fungus Aspergillus fumigatus. Molecules. 2015;20:1424–1433. doi: 10.3390/molecules20011424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Linh TM, Mai NC, Hoe PT, Ngoc NT, Thao PT, Ban NK, Van NT. Development of a cell suspension culture system for promoting alkaloid and vinca alkaloid biosynthesis using endophytic fungi isolated from local Catharanthus roseus. Plants. 2021;10(4):672. doi: 10.3390/plants10040672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Liu L, Liu S, Niu S, Guo L, Chen X, Che Y. Isoprenylated chromone derivatives from the plant endophytic fungus Pestalotiopsis fici. J Nat Prod. 2009;72(8):1482–14826. doi: 10.1021/np900308s. [DOI] [PubMed] [Google Scholar]
  161. Liu X, Dou G, Ma Y. Potential of endophytes from medicinal plants for biocontrol and plant growth promotion. J Gen Plant Pathol. 2016;82(3):165–173. doi: 10.1007/s10327-016-0648-9. [DOI] [Google Scholar]
  162. Liu J, Wang X, Pu H, Liu S, Kan J, Jin C. Recent advances in endophytic exopolysaccharides: production, structural characterization, physiological role and biological activity. Carbohydr Polym. 2017;157:1113–1124. doi: 10.1016/j.carbpol.2016.10.084. [DOI] [PubMed] [Google Scholar]
  163. Liu Q, Johnson LJ, Applegate ER, Arfmann K, Jauregui R, Larking A, Mace WJ, Maclean P, Walker T, Johnson RD. Identification of genetic diversity, pyrrocidine-producing strains and transmission modes of endophytic Sarocladium zeae fungi from Zea crops. Microorganisms. 2022;10(7):1415. doi: 10.3390/microorganisms10071415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Lubna AS, Hamayun M, Gul H, Lee IJ, Hussain A. Aspergillus niger CSR3 regulates plant endogenous hormones and secondary metabolites by producing gibberellins and indoleacetic acid. J Plant Interact. 2018;13:100–111. doi: 10.1080/17429145.2018.1436199. [DOI] [Google Scholar]
  165. Lubna KMA, Asaf S, Jan R, Waqas M, Kim KM, Lee IJ. Endophytic fungus Bipolaris sp. CSL-1 induces salt tolerance in Glycine max. L via modulating its endogenous hormones, anti-oxidative system and gene expression. J Plant Interact. 2022;17(1):319–332. doi: 10.1080/17429145.2022.2036836. [DOI] [Google Scholar]
  166. Lykholat YV, Khromykh NO, Didur OO, Drehval OA, Sklyar TV, Anishchenko AO. Chaenomeles speciosa fruit endophytic fungi isolation and characterization of their anti-microbial activity and the secondary metabolites composition. Beni-Seuf Univ Basic J Appl Sci. 2021;10(1):1. doi: 10.1186/s43088-021-00171-2. [DOI] [Google Scholar]
  167. Ma Y, Prasad MN, Rajkumar M, Freitas H. Plant growth promoting rhizobacteria and endophytes accelerate phytoremediation of metalliferous soils. Biotechnol Adv. 2011;29(2):248–258. doi: 10.1016/j.biotechadv.2010.12.001. [DOI] [PubMed] [Google Scholar]
  168. Macías-Rubalcava ML, Sánchez-Fernández RE. Secondary metabolites of endophytic Xylaria species with potential applications in medicine and agriculture. World J Microbiol Biotechnol. 2017;33(1):15. doi: 10.1007/s11274-016-2174-5. [DOI] [PubMed] [Google Scholar]
  169. Madasi A, Ajmeera A, Renuka G, Vemireddy B, Vanteru KR. Production of extracellular enzymes, anti-microbial and other agriculturally important metabolites by fungal endophytes of Litsea glutinosa (Lour.) CB Rob, a medicinal plant. J Pure Appl Microbiol. 2021;15(4):2317–2329. doi: 10.22207/JPAM.15.4.5. [DOI] [Google Scholar]
  170. Maehara S, Simanjuntak P, Ohashi K, Shibuya H. Composition of endophytic fungi living in Cinchona ledgeriana (Rubiaceae) J Nat Med. 2010;64(2):227–230. doi: 10.1007/s11418-009-0380-2. [DOI] [PubMed] [Google Scholar]
  171. Magotra A, Kumar M, Kushwaha M, Awasthi P, Raina C, Gupta AP, Shah BA, Gandhi SG, Chaubey A. Epigenetic modifier induced enhancement of fumiquinazoline C production in Aspergillus fumigatus (GA-L7): an endophytic fungus from Grewia asiatica L. AMB Express. 2017;7(1):1. doi: 10.1186/s13568-017-0343-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Mahapatra S, Banerjee D. Structural elucidation and bioactivity of a novel exopolysaccharide from endophytic Fusarium solani SD5. Carbohydr Polym. 2012;90(1):683–689. doi: 10.1016/j.carbpol.2012.05.097. [DOI] [PubMed] [Google Scholar]
  173. Mahapatra S, Banerjee D. Evaluation of in vitro anti-oxidant potency of exopolysaccharide from endophytic Fusarium solani SD5. Int J Biol Macromol. 2013;53(2):62–66. doi: 10.1016/j.ijbiomac.2012.11.003. [DOI] [PubMed] [Google Scholar]
  174. Maharjan S, Lee SB, Kim GJ, Cho SJ, Nam JW, Chin J, Choi H. Isolation of unstable isomers of lucilactaene and evaluation of anti-inflammatory activity of secondary metabolites produced by the endophytic fungus Fusarium sp. QF001 from the roots of Scutellaria baicalensis. Molecules. 2020;25(4):923. doi: 10.3390/molecules25040923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Mani VM, Gnana Soundari AP, Karthiyaini D, Preethi K. Bioprospecting endophytic fungi and their metabolites from medicinal tree Aegle marmelos in Western Ghats, India. Mycobiology. 2015;43:303–310. doi: 10.5941/MYCO.2015.43.3.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Mapari SA, Meyer AS, Thrane U. Photostability of natural orange−red and yellow fungal pigments in liquid food model systems. J Agric Food Chem. 2009;57(14):6253–6261. doi: 10.1021/jf900113q. [DOI] [PubMed] [Google Scholar]
  177. Marín F, Navarrete H, Narvaez-Trujillo A. Total petroleum hydrocarbon degradation by endophytic fungi from the Ecuadorian Amazon. Adv Appl Microbiol. 2018;8(12):1029–1053. doi: 10.4236/aim.2018.812070. [DOI] [Google Scholar]
  178. Mathur P, Ghosh U, Chetani R, Chaturvedi P, Sharma C, Bhatnagar P. Enzymatic bioprospection of endophytic Aspergillus niger isolated from Albizia lebbeck (L.) Benth. South Afr J Bot. 2022;148:580–587. doi: 10.1016/j.sajb.2022.05.033. [DOI] [Google Scholar]
  179. Mehmood A, Hussain A, Irshad M, Hamayun M, Iqbal A, Rahman A, Tawab A, Ahmad A, Ayaz S. Cinnamic acid as an inhibitor of growth, favonoids exudation and endophytic fungus colonization in maize root. Plant Physiol Biochem. 2019;135:61–68. doi: 10.1016/j.plaphy.2018.11.029. [DOI] [PubMed] [Google Scholar]
  180. Ming Q, Su C, Zheng C, Jia M, Zhang Q, Zhang H, Rahman K, Han T, Qin L. Elicitors from the endophytic fungus Trichoderma atroviride promote Salvia miltiorrhiza hairy root growth and tanshinone biosynthesis. J Exp Bot. 2013;64:5687–5694. doi: 10.1093/jxb/ert342. [DOI] [PubMed] [Google Scholar]
  181. Mir RA, Kaushik SP, Chowdery RA, Anuradha M. Elicitation of forskolin in cultures of Rhizactonia bataticola-a phytochemical synthesizing endophytic fungi. Int J Pharm Pharma Sci. 2015;7:185–189. [Google Scholar]
  182. Mishra VK, Passari AK, Chandra P, Leo VV, Kumar B, Uthandi S, et al. Determination and production of anti-microbial compounds by Aspergillus clavatonanicus strain MJ31, an endophytic fungus from Mirabilis jalapa L. using UPLC–ESI–MS/MS and TD–GC–MS analysis. PLoS ONE. 2017;12:e0186234. doi: 10.1371/journal.pone.0186234. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  183. Mochahari D, Kharnaior S, Sen S, Thomas SC. Isolation of endophytic fungi from juvenile Aquilaria malaccensis and their anti-microbial properties. J Trop for Sci. 2020;32:97–104. doi: 10.2307/26872825. [DOI] [Google Scholar]
  184. Mohamed H, Ebrahim W, El-Neketi M, Awad MF, Zhang H, Zhang Y, Song Y. In vitro phytobiological investigation of bioactive secondary metabolites from the malus domestica-derived endophytic fungus Aspergillus tubingensis strain AN103. Molecules. 2022;27(12):3762. doi: 10.3390/molecules27123762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Monggoot S, Popluechai S, Gentekaki E, Pripdeevech P. Fungal endophytes: an alternative source for production of volatile compounds from agarwood oil of Aquilaria subintegra. Microb Ecol. 2017;74(1):54–61. doi: 10.1007/s00248-016-0908-4. [DOI] [PubMed] [Google Scholar]
  186. Moscovici M. Present and future medical applications of microbial exopolysaccharides. Front Microbiol. 2015;6:1012. doi: 10.3389/fmicb.2015.01012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Motoyama T, Yun CS, Osada H. Biosynthesis and biological function of secondary metabolites of the rice blast fungus Pyricularia oryzae. J Ind Microbiol. 2021;48:9–10. doi: 10.1093/jimb/kuab058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Mózsik L, Pohl C, Meyer V, Bovenberg RA, Nygård Y, Driessen AJ. Modular synthetic biology toolkit for filamentous fungi. ACS Synth Biol. 2021;10(11):2850–2861. doi: 10.1021/acssynbio.1c00260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Mózsik L, Iacovelli R, Bovenberg RAL, Driessen AJM. Transcriptional activation of biosynthetic gene clusters in filamentous fungi. Front Bioeng Biotechnol. 2022;10:901037. doi: 10.3389/fbioe.2022.901037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Musetti R, Vecchione A, Stringher L, Borselli S, Zulini L, Marzani C, D’Ambrosio M, Di TL, Pertot I. Inhibition of sporulation and ultrastructural alterations of grapevine downy mildew by the endophytic fungus Alternaria alternata. Phytopathology. 2006;96(7):689–698. doi: 10.1094/PHYTO-96-0689. [DOI] [PubMed] [Google Scholar]
  191. Nagarajan A, Thirunavuk N, Suryanaray TS, Gummadi SN. Screening and isolation of novel glutaminase free L-asparaginase from fungal endophytes. Res J Microbiol. 2014;9(4):163–176. doi: 10.3923/jm.2014.163.176. [DOI] [Google Scholar]
  192. Nath R, Sharma GD, Barooah M. Efficiency of tricalcium phosphate solubilization by two different endophytic Penicillium sp. isolated from tea (Camellia sinensis L.) Eur J Exp Biol. 2012;2(4):1354–1358. [Google Scholar]
  193. Nath A, Chattopadhyay A, Joshi S. Biological activity of endophytic fungi of Rauwolfia serpentina Benth: an ethnomedicinal plant used in folk medicines in Northeast India. Proc Natl Acad Sci. 2015;85(1):233–240. doi: 10.1007/s40011-013-0184-8. [DOI] [Google Scholar]
  194. Neha C, Ram P, Ajit V. Plant promoting activities of fungal endophytes associated with tomato roots from Central Himalaya, India and their interaction with Piriformospora indica. Int J Pharm Bio Sci. 2015;6(1):333–343. [Google Scholar]
  195. Netala VR, Bethu MS, Pushpalatha B, Baki VB, Aishwarya S, Rao JV, Tartte V. Biogenesis of silver nanoparticles using endophytic fungus Pestalotiopsis microspora and evaluation of their antioxidant and anticancer activities. Int J Nanomedicine. 2016;11:5683. doi: 10.2147/IJN.S112857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Netala VR, Kotakadi VS, Bobbu P, Gaddam SA, Tartte V. Endophytic fungal isolate mediated biosynthesis of silver nanoparticles and their free radical scavenging activity and anti-microbial studies. 3 Biotech. 2016;6:1–9. doi: 10.1007/s13205-016-0433-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Nischitha R, Shivanna MB. Screening of secondary metabolites and anti-oxidant potential of endophytic fungus Penicillium citrinum and host Digitaria bicornis by spectrophotometric and electrochemical methods. Arch Microbiol. 2022;204(4):1–3. doi: 10.1007/s00203-022-02795-z. [DOI] [PubMed] [Google Scholar]
  198. Notarte KIR, Devanadera MKP, Mayor ABR, Cada MCA, Pecundo MH, Macabeo APG. Toxicity, anti-bacterial, and anti-oxidant activities of fungal endophytes Colletotrichum and Nigrospora spp. Isolated from Uvaria grandiflora. Phipp J Sci. 2019;148:503–510. [Google Scholar]
  199. Nwe N, Furuike T, Tamura H. The mechanical and biological properties of chitosan scaffolds for tissue regeneration templates are significantly enhanced by chitosan from Gongronella butleri. Materials. 2009;2(2):374–398. doi: 10.3390/ma2020374. [DOI] [Google Scholar]
  200. Oberhofer M, Malfent F, Zehl M, Urban E, Wackerlig J, Reznicek G, Vignolle GA, Rückert C, Busche T, Wibberg D, Zotchev SB. Biosynthetic potential of the endophytic fungus Helotiales sp. BL73 revealed via compound identification and genome mining. Appl Environ Microbiol. 2022;88(6):e02510–21. doi: 10.1128/aem.02510-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Onn ML, Lim PT, Mujahid A, Proksch P, Müller M. Initial screening of mangrove endophytic fungi for anti-microbial compounds and heavy metal biosorption potential. Sains Malaysiana. 2016;45(7):1063–1071. [Google Scholar]
  202. Orlandelli RC, Silva da Silva ML, Vasconcelos AFD, Almeida IVD, Vicentini VEP, Prieto A, Hernandez MDD, Azevedo JLD, Pamphile JA. β-(1→3,1→6)-D-glucans produced by Diaporthe sp. endophytes: purification, chemical characterization and antiproliferative activity against MCF-7 and HepG2-C3A cells. Int J Biol Macromol. 2017;94(Pt A):431–437. doi: 10.1016/j.ijbiomac.2016.10.048. [DOI] [PubMed] [Google Scholar]
  203. Ownley BH, Griffin MR, Klingeman WE, Gwinn KD, Moulton JK, Pereira RM. Beauveria bassiana: endophytic colonization and plant disease control. J Invertebr Pathol. 2008;98(3):267–270. doi: 10.1016/j.jip.2008.01.010. [DOI] [PubMed] [Google Scholar]
  204. Pádua APSLD, Freire KTLDS, Oliveira TGLD, Silva LFD, Araújo-Magalhães GR, Agamez-Montalvo GS, Silva IRD, Bezerra JDP, Souza-Motta CMD. Fungal endophyte diversity in the leaves of the medicinal plant Myracrodruon urundeuva in a Brazilian dry tropical forest and their capacity to produce L-asparaginase. Acta Bot Bras. 2019;33:39–49. doi: 10.1590/0102-33062018abb0108. [DOI] [Google Scholar]
  205. Palanichamy P, Kannan S, Murugan D, Alagusundaram P, Marudhamuthu M. Purification, crystallization and anti-cancer activity evaluation of the compound alternariolmethyl ether from endophytic fungi Alternaria alternata. J Appl Microbiol. 2019;127:1468–1478. doi: 10.1111/jam.14410. [DOI] [PubMed] [Google Scholar]
  206. Palem PPC, Kuriakose GC, Jayabaskaran C. An endophytic fungus, Talaromyces radicus, isolated from Catharanthus roseus, produces vincristine and vinblastine, which induce apoptotic cell death. PLoS ONE. 2015;10(12):0144476. doi: 10.1371/journal.pone.0153111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Pan BF, Su X, Hu B, Yang N, Chen Q, Wu W. Fusarium redolens 6WBY3, an endophytic fungus isolated from Fritillaria unibracteata var. wabuensis, produces peimisine and imperialine-3β-D-glucoside. Fitoterapia. 2015;103:213–221. doi: 10.1016/j.fitote.2015.04.006. [DOI] [PubMed] [Google Scholar]
  208. Pan F, Su TJ, Cai SM, Wu W. Fungal endophyte derived Fritillaria unibracteata var. wabuensis: diversity, anti-oxidant capacities in vitro and relations to phenolic, flavonoid or saponin compounds. Sci Rep. 2017;7:42008. doi: 10.1038/srep42008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Pandey SS, Singh S, Babu CS, 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):1–4. doi: 10.1038/srep26583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Pandey SS, Singh S, Babu CV, 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;243:1097–1114. doi: 10.1007/s00425-016-2467-9. [DOI] [PubMed] [Google Scholar]
  211. Pappas ML, Liapoura M, Papantoniou D, Avramidou M, Kavroulakis N, Weinhold A, Broufas GD, Papadopoulou KK. The beneficial endophytic fungus Fusarium solani strain K alters tomato responses against spider mites to the benefit of the plant. Front Plant Sci. 2018;9:1603. doi: 10.3389/fpls.2018.01603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Parthasarathy R, Sathiyabama M. Gymnemagenin-producing endophytic fungus isolated from a medicinal plant Gymnema sylvestre R.Br. Appl Biochem Biotechnol. 2014;172(6):3141–3152. doi: 10.1007/s12010-014-0754-0. [DOI] [PubMed] [Google Scholar]
  213. Parthasarathy R, Sathiyabama M. Lovastatin-producing endophytic fungus isolated from a medicinal plant Solanum xanthocarpum. Nat Prod Res. 2015;29(24):2282–2286. doi: 10.1080/14786419.2015.1016938. [DOI] [PubMed] [Google Scholar]
  214. Pasha C, Reddy G. Nutritional and medicinal improvement of black tea by yeast fermentation. Food Chem. 2005;89(3):449–453. doi: 10.1016/j.foodchem.2004.02.054. [DOI] [Google Scholar]
  215. Peng W, Ming QL, Zhai X, Zhang Q, Rahman K, Wu SJ, Qin LP, Han T. Polysaccharide fraction extracted from endophytic fungus Trichoderma atroviride D16 has an influence on the proteomics profile of the Salvia miltiorrhiza hairy roots. Biomolecules. 2019;9:415. doi: 10.3390/biom9090415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Pietro-Souza W, de Campos Pereira F, Mello IS, Stachack FF, Terezo AJ, da Cunha CN, White JF, Li H, Soares MA. Mercury resistance and bioremediation mediated by endophytic fungi. Chemosphere. 2020;240:124874. doi: 10.1016/j.chemosphere.2019.124874. [DOI] [PubMed] [Google Scholar]
  217. Pinheiro EA, Pina JR, Feitosa AO, Carvalho JM, Borges FC, Marinho PS, Marinho AM. Bioprospecting of anti-microbial activity of extracts of endophytic fungi from Bauhinia guianensis. Rev Argent Microbiol. 2017;49(1):3–6. doi: 10.1016/j.ram.2016.08.005. [DOI] [PubMed] [Google Scholar]
  218. Pohl C, Kiel JAKW, Driessen AJM, Bovenberg RAL, Nygård Y. CRISPR/Cas9 based genome editing of Penicillium chrysogenum. ACS Synth Biol. 2016;5:754–764. doi: 10.1021/acssynbio.6b00082. [DOI] [PubMed] [Google Scholar]
  219. Polturak G, Osbourn A. The emerging role of biosynthetic gene clusters in plant defense and plant interactions. PLoS Pathog. 2021;17(7):e1009698. doi: 10.1371/journal.ppat.1009698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Prabukumar S, Rajkuberan C, Ravindran K, Sivaramakrishnan S. Isolation and characterization of endophytic fungi from medicinal plant Crescentia cujete L. and their anti-bacterial, anti-oxidant and anti-cancer properties. Int J Pharm Pharm Sci. 2015;7(11):316–321. [Google Scholar]
  221. Praptiwi P, Ilyas M, Palupi KD, Fathoni A, Evana E, Raunsai MM, Agusta A. Rapid screening of anti-bacterial and anti-oxidant metabolites from endophytic fungi isolated from Papuacedrus papuana by TLC-bioautography. J Kim Dan Kemasan. 2021;43(2):110–116. doi: 10.24817/jkk.v43i2.6980. [DOI] [Google Scholar]
  222. Prihantini AI, Tachibana S. Anti-oxidant compounds produced by Pseudocercospora sp. ESL 02, an endophytic fungus isolated from Elaeocarpus sylvestris. Asian Pac J Trop Biomed. 2017;7(2):110–115. doi: 10.1016/j.apjtb.2016.11.020. [DOI] [Google Scholar]
  223. Priyadharsini P, Muthukumar T. The root endophytic fungus Curvularia geniculata from Parthenium hysterophorus roots improves plant growth through phosphate solubilization and phytohormone production. Fungal Ecol. 2017;27:69–77. doi: 10.1016/j.funeco.2017.02.007. [DOI] [Google Scholar]
  224. Pu X, Qu X, Chen F, Bao J, Zhang G, Luo Y. Camptothecin producing endophytic fungus Trichoderma atroviride LY357: isolation, identification, and fermentation conditions optimization for camptothecin production. Appl Microbiol Biotechnol. 2013;97(21):9365–9375. doi: 10.1007/s00253-013-5163-8. [DOI] [PubMed] [Google Scholar]
  225. Puri SC, Nazir A, Chawla R, Arora R, Riyaz-ul-Hasan S, Amna T, Ahmed B, Verma V, Singh S, Sagar R, Sharma A. The endophytic fungus Trametes hirsuta as a novel alternative source of podophyllotoxin and related aryl tetralin lignans. J Biotechnol. 2006;122(4):494–510. doi: 10.1016/j.jbiotec.2005.10.015. [DOI] [PubMed] [Google Scholar]
  226. Qadri M, Nalli Y, Jain SK, Chaubey A, Ali A, Strobel GA, Vishwakarma RA, Riyaz-Ul-Hassan S. An insight into the secondary metabolism of Muscodor yucatanensis: small-molecule epigenetic modifiers induce expression of secondary metabolism-related genes and production of new metabolites in the endophyte. Microb Ecol. 2017;73(4):954–965. doi: 10.1007/s00248-016-0901-y. [DOI] [PubMed] [Google Scholar]
  227. Raheman F, Deshmukh S, Ingle A, Gade A, Rai M. Silver nanoparticles: novel antimicrobial agent synthesized from an endophytic fungus Pestalotia sp. isolated from leaves of Syzygium cumini (L) Nano Biomed Eng. 2011;3(3):174–178. doi: 10.5101/nbe.v3i3.p174-178. [DOI] [Google Scholar]
  228. Rajulu MBG, Thirunavukkarasu N, Suryanarayanan TS, Ravishankar JP, Gueddari NEE, Moerschbacher BM. Chitinolytic enzymes from endophytic fungi. Fungal Divers. 2011;47(1):43–53. doi: 10.1007/s13225-010-0071-z. [DOI] [Google Scholar]
  229. Ramírez-Villalobos JM, Gomez-Flores R, Velázquez-Flores PV, Morán-Santibáñez KS, Tamez-Guerra P, Pérez-González O, de la Garza-Ramos MA, Rodríguez-Padilla C, Romo-Sáenz CI. Effect of culture conditions of Lophocereus marginatus endophytic fungi on yield and anticancer and antioxidant activities. Int J Env Res Pub Heal. 2023;20(5):3948. doi: 10.3390/ijerph20053948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Rashmi M, Venkateswara SV. Secondary metabolite production by endophytic fungi: the gene clusters, nature, and expression. In: Jha S, editor. Endophytes and secondary metabolites. New York: Springer; 2019. pp. 475–490. [Google Scholar]
  231. Rauf M, Ur-Rahman A, Arif M, Gul H, Ud-Din A, Hamayun M, Lee IJ. Immunomodulatory molecular mechanisms of Luffa cylindrica for downy mildews resistance induced by growth-promoting endophytic fungi. J Fungi. 2022;8(7):689. doi: 10.3390/jof8070689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Ravi P, Somu P, Acharya D, Gomez LA, Thathapudi JJ, Ramachandra YL, Rudraiah SB, Isaq M, Karua CS, Arifullah M, Poojari CC. Isolation and phytochemical screening of endophytic fungi isolated from medicinal plant Mappia foetida and evaluation of its in vitro cytotoxicity in cancer. Appl Biochem Biotechnol. 2022 doi: 10.1007/s12010-022-03929-1. [DOI] [PubMed] [Google Scholar]
  233. Ray T, Pandey SS, Pandey A, Srivastava M, Shanker K, Kalra A. Endophytic consortium with diverse gene-regulating capabilities of benzylisoquinoline alkaloids biosynthetic pathway can enhance endogenous morphine biosynthesis in Papaver somniferum. Front Microbiol. 2019;10:925. doi: 10.3389/fmicb.2019.00925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Ren CG, Dai CC. Jasmonic acid is involved in the signaling pathway for fungal endophyte-induced volatile oil accumulation of Atractylodes lancea plantlets. BMC Plant Biol. 2012;12:1–1. doi: 10.1186/1471-2229-12-128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Ren H, Shi C, Zhao H. Computational tools for discovering and engineering natural product biosynthetic pathways. Iscience. 2020;23(1):100795. doi: 10.1016/j.isci.2019.100795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Rinu K, Sati P, Pandey A. Trichoderma gamsii (NFCCI 2177): a newly isolated endophytic, psychrotolerant, plant growth promoting, and antagonistic fungal strain. J Basic Microbiol. 2014;54(5):408–417. doi: 10.1002/jobm.201200579. [DOI] [PubMed] [Google Scholar]
  237. Rodrigues-Corrêa KC, Fett-Neto AG. Abiotic stresses and non-protein amino acids in plants. CRC Crit Rev Plant Sci. 2019;38(5–6):411–430. doi: 10.1080/07352689.2019.1707944. [DOI] [Google Scholar]
  238. Rodriguez Estrada AE, Jonkers W, Corby Kistler H, May G. Interactions between Fusarium verticillioides, Ustilago maydis, and Zea mays: an endophyte, a pathogen, and their shared plant host. Fungal Genet Biol. 2012 doi: 10.1016/j.fgb.2012.05.001. [DOI] [PubMed] [Google Scholar]
  239. Rojas EC, Jensen B, Jørgensen HJ, Latz MA, Esteban P, Collinge DB. The fungal endophyte Penicillium olsonii ML37 reduces Fusarium head blight by local induced resistance in wheat spikes. J Fungus. 2022;8(4):345. doi: 10.3390/jof8040345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Rokas A, Mead ME, Steenwyk JL, Raja HA, Oberlies NH. Biosynthetic gene clusters and the evolution of fungal chemodiversity. Nat Prod Rep. 2020;37(7):868–878. doi: 10.1039/C9NP00045C. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Rondot Y, Reineke A. Endophytic Beauveria bassiana activates expression of defence genes in grapevine and prevents infections by grapevine downy mildew Plasmopara viticola. Plant Pathol. 2019;68(9):1719–1731. doi: 10.1111/ppa.13089. [DOI] [Google Scholar]
  242. Sagita R, Quax WJ, Haslinger K. Current state and future directions of genetics and genomics of endophytic fungi for bioprospecting efforts. Front Bioeng Biotechnol. 2021;15(9):649906. doi: 10.3389/fbioe.2021.649906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Samaga PV, Rai VR. Diversity and bioactive potential of endophytic fungi from Nothapodytes foetida, Hypericum mysorense and Hypericum japonicum collected from Western Ghats of India. Annal Microbiol. 2016;66(1):229–244. doi: 10.1007/s13213-015-1099-9. [DOI] [Google Scholar]
  244. Santos TF, dos Santos CC, de Almeida MA, Delforno TP, Duarte IC. Endophytic fungi isolated from Brazilian medicinal plants as potential producers of anti-oxidants and their relations with anti-inflammatory activity. 3 Biotech. 2020;10:223. doi: 10.1007/s13205-020-02211-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Santosfo F, Fill TP, Nakamura J, Monteiro MR, Rodriguesfo E. Endophytic fungi as a source of biofuel precursors. J Microbiol Biotechnol. 2011;21(7):728–733. doi: 10.4014/jmb.1010.10052. [DOI] [PubMed] [Google Scholar]
  246. Selim KA, Elkhateeb WA, Tawila AM, El-Beih AA, Abdel-Rahman TM, El-Diwany AI, Ahmed EF. Antiviral and anti-oxidant potential of fungal endophytes of Egyptian medicinal plants. Fermentation. 2018;4(3):49. doi: 10.3390/fermentation4030049. [DOI] [Google Scholar]
  247. Senthivelan T, Kanagaraj J, Panda RC. Recent trends in fungal laccase for various industrial applications: an eco-friendly approach-a review. Biotechnol Bioprocess Eng. 2016;21(1):19–38. doi: 10.1007/s12257-015-0278-7. [DOI] [Google Scholar]
  248. Shaw JJ, Spakowicz DJ, Dalal RS, Davis JH, Lehr NA, Dunican BF, Orellana EA, Narváez-Trujillo A, Strobel SA. Biosynthesis and genomic analysis of medium-chain hydrocarbon production by the endophytic fungal isolate Nigrograna mackinnonii E5202H. Appl Microbiol Biotechnol. 2015;99(8):3715–3728. doi: 10.1007/s00253-014-6206-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Sheik S, Chandrashekar KR. Fungal endophytes of an endemic plant Humboldtia brunonis Wall. of Western Ghats (India) and their anti-microbial and DPPH radical scavenging potentiality. Oriental Pharm Exp Med. 2018;18:115–125. doi: 10.1007/s13596-018-0297-y. [DOI] [Google Scholar]
  250. Shi JL, Liu C, Liu L, Yang B, Zhang Y. Structure identification and fermentation characteristics of pinoresinol diglucoside produced by Phomopsis sp. isolated from Eucommia ulmoides Oliv. Appl Microbiol Biotechnol. 2012;93(4):1475–1483. doi: 10.1007/s00253-011-3613-8. [DOI] [PubMed] [Google Scholar]
  251. Shi Y, Xie H, Cao L, Zhang R, Xu Z, Wang Z, Deng Z. Effects of Cd- and Pb-resistant endophytic fungi on growth and phytoextraction of Brassica napus in metal-contaminated soils. Environ Sci Pollut Res. 2017;24(1):417–426. doi: 10.1007/s11356-016-7693-y. [DOI] [PubMed] [Google Scholar]
  252. Shweta S, Gurumurthy BR, Ravikanth G, Ramanan US, Shivanna MB. Endophytic fungi from Miquelia dentata Bedd., produce the anti-cancer alkaloid, camptothecine. Phytomedicine. 2013;20(3–4):337–342. doi: 10.1016/j.phymed.2012.11.015. [DOI] [PubMed] [Google Scholar]
  253. Siddaiah CN, Satyanarayana NR, Mudili V, Gupta VK, Gurunathan S, Rangappa S, Huntrike SS, Srivastava RK. Elicitation of resistance and associated defense responses in Trichoderma hamatum induced protection against pearl millet downy mildew pathogen. Sci Rep. 2017;7:43991. doi: 10.1038/srep43991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Siebatcheu EC, Wetadieu D, Youassi Youassi O, Bedine Boat MA, Bedane KG, Tchameni NS, Sameza ML. Secondary metabolites from an endophytic fungus Trichoderma erinaceum with anti-microbial activity towards Pythium ultimum. Nat Prod Res. 2022;10:1–6. doi: 10.1080/14786419.2022.2075360. [DOI] [PubMed] [Google Scholar]
  255. Silva AA, Polonio JC, Bulla AM, Polli AD, Castro JC, Soares LC, Oliveira-Junior VA, Vicentini VE, Oliveira AJ, Gonçalves JE, Gonçalves RA. Anti-microbial and anti-oxidant activities of secondary metabolites from endophytic fungus Botryosphaeria fabicerciana (MGN23-3) associated to Morus nigra L. Nat Prod Res. 2021;24:1–5. doi: 10.1080/14786419.2021.1947272. [DOI] [PubMed] [Google Scholar]
  256. 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):1128–1142. doi: 10.1111/jam.14546. [DOI] [PubMed] [Google Scholar]
  257. Singh SK, Patra B, Paul P, Liu Y, Pattanaik S, Yuan L. Revisiting the ORCA gene cluster that regulates terpenoid indole alkaloid biosynthesis in Catharanthus roseus. Plant Sci. 2020;293:110408. doi: 10.1016/j.plantsci.2020.110408. [DOI] [PubMed] [Google Scholar]
  258. Soleimani M, Afyuni M, Hajabbasi MA, Nourbakhsh F, Sabzalian MR, Christensen JH. Phytoremediation of an aged petroleum contaminated soil using endophyte infected and non-infected grasses. Chemosphere. 2010;81(9):1084–1090. doi: 10.1016/j.chemosphere.2010.09.034. [DOI] [PubMed] [Google Scholar]
  259. Soliman SS, Raizada MN. Interactions between co-habitating fungi elicit synthesis of taxol from an endophytic fungus in host taxus plants. Front Microbiol. 2013;4:3. doi: 10.3389/fmicb.2013.00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Soliman SSM, Raizada MN. Interactions between co-habitating fungi elicit synthesis of taxol from an endophytic fungus in host Taxus Plants. Front Microbiol. 2013;4(1):3. doi: 10.3389/fmicb.2013.00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Soliman SSM, Greenwood JS, Bombarely A, Mueller LA, Tsao R, Mosser DD, Raizada MN. An endophyte constructs fungicide-containing extracellular barriers forits host plant. Curr Biol. 2015;25(19):2570–2576. doi: 10.1016/j.cub.2015.08.027. [DOI] [PubMed] [Google Scholar]
  262. Spagnoletti FN, Tobar NE, Di Pardo AF, Chiocchio VM, Lavado RS. Dark septate endophytes present different potential to solubilize calcium, iron and aluminum phosphates. Appl Soil Ecol. 2017;111:25–32. doi: 10.1016/j.apsoil.2016.11.010. [DOI] [Google Scholar]
  263. Staropoli A, Iacomino G, De Cicco P, Woo SL, Di Costanzo L, Vinale F. Induced secondary metabolites of the beneficial fungus Trichoderma harzianum M10 through OSMAC approach. Chem Biol Technol Agric. 2023;10(1):28. doi: 10.1186/s40538-023-00383-x. [DOI] [Google Scholar]
  264. Stierle A, Strobel G, Stierle D. Taxol and taxane production by Taxomyces andreanae, an endophytic fungus of Pacific yew. Science. 1993;260(5105):214–216. doi: 10.1126/science.8097061’. [DOI] [PubMed] [Google Scholar]
  265. Strobel G, Knighton B, Kluck K, Ren Y, Livinghouse T, Griffin M, Spakowicz D, Sears J. The production of myco-diesel hydrocarbons and their derivatives by the endophytic fungus Gliocladium roseum (NRRL 50072) Microbiology. 2008;154(11):3319–3328. doi: 10.1099/mic.0.30824-0. [DOI] [PubMed] [Google Scholar]
  266. Strobel GA, Pliam NB (1997) Immuno suppressants diterpene compounds. Google Patents.
  267. Stroe MC, Netzker T, Scherlach K, Krüger T, Hertweck C, Valiante V, Brakhage AA. Targeted induction of a silent fungal gene cluster encoding the bacteria-specific germination inhibitor fumigermin. Elife. 2020;9:e52541. doi: 10.7554/eLife.52541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Sun C, Johnson JM, Cai D, Sherameti I, Oelmüller R, Lou B. Piriformospora indica confers drought tolerance in Chinese cabbage leaves by stimulating anti-oxidant enzymes, the expression of drought-related genes and the plastid-localized CAS protein. J Plant Physiol. 2010;167(12):1009–1017. doi: 10.1016/j.jplph.2010.02.013. [DOI] [PubMed] [Google Scholar]
  269. Suradkar K, Hande D. Characterization of bioactive secondary metabolites of fungal endophytes from Melghat Forest in Maharashtra, India. In: Singh J, Meshram V, Gupta M, editors. Bioactive natural products in drug discovery. Singapore: Springer; 2020. pp. 599–607. [Google Scholar]
  270. Surono NK. The dark septate endophytic fungus Phialocephala fortinii is a potential decomposer of soil organic compounds andapromoter of Asparagus officinalis growth. Fungal Ecol. 2017;28:1–10. doi: 10.1016/j.funeco.2017.04.001. [DOI] [Google Scholar]
  271. Suryanarayanan TS, Thirunavukkarasu N, Govindarajulu MB, Gopalan V. Fungal endophytes: an untapped source of biocatalysts. Fungal Divers. 2012;54(1):19–30. doi: 10.1007/s13225-012-0168-7. [DOI] [Google Scholar]
  272. Suryavamshi G, Shivanna MB. Diversity and anti-bacterial activity of endophytic fungi in Memecylon umbellatum Burm. F.-a medicinal plant in the Western Ghats of Karnataka, India. Indian J Ecol. 2020;47:171–180. [Google Scholar]
  273. Syarifah S, Elfita E, Widjajanti H, Setiawan A, Kurniawati AR. Diversity of endophytic fungi from the root bark of Syzygium zeylanicum, and the anti-bacterial activity of fungal extracts, and secondary metabolite. Biodivers J Biol Divers. 2021 doi: 10.13057/biodiv/d221051. [DOI] [Google Scholar]
  274. 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. [Google Scholar]
  275. Tao J, Bai X, Zeng M, Li M, Hu Z, Hua Y, Zhang H. Whole-genome sequence analysis of an endophytic fungus Alternaria sp. SPS-2 and its biosynthetic potential of bioactive secondary metabolites. Microorganisms. 2022;10(9):1789. doi: 10.3390/microorganisms10091789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Terhonen E, Sipari N, Asiegbu FO. Inhibition of phytopathogens by fungal root endophytes of Norway spruce. Biol Control. 2016;99:53–63. doi: 10.1016/j.biocontrol.2016.04.006. [DOI] [Google Scholar]
  277. Theobald S, Vesth TC, Rendsvig JK, Nielsen KF, Riley R, de Abreu LM, Salamov A, Frisvad JC, Larsen TO, Andersen MR, Hoof JB. Uncovering secondary metabolite evolution and biosynthesis using gene cluster networks and genetic dereplication. Sci Rep. 2018;8(1):1–2. doi: 10.1038/s41598-018-36561-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Thirunavukkarasu N, Jahnes B, Broadstock A, Rajulu MBG, Murali TS, Gopalan V, Suryanarayanan TS. Screening marine-derived endophytic fungi for xylan-degrading enzymes. Curr Sci. 2015;109(1):112–120. [Google Scholar]
  279. Tiwari P, Bae H. Horizontal gene transfer and endophytes: an implication for the acquisition of novel traits. Plants. 2020;9(3):305. doi: 10.3390/plants9030305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Tiwari P, Bae H. Endophytic fungi: key insights, emerging prospects, and challenges in natural product drug discovery. Microorganisms. 2022;10(2):360. doi: 10.3390/microorganisms10020360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Toghueo RMK, Zabalgogeazcoa I, de Aldana BV, Boyom FF. Enzymatic activity of endophytic fungi from the medicinal plants Terminalia catappa, Terminalia mantaly and Cananga odorata. S Afr J Bot. 2017;109:146–153. doi: 10.1016/j.sajb.2016.12.021. [DOI] [Google Scholar]
  282. Toppo P, Subba R, Roy K, Mukherjee S, Mathur P. Elucidating the strategies for isolation of endophytic fungi and their functional attributes for the regulation of plant growth and resilience to stress. J Plant Growth Regul. 2022 doi: 10.1007/s00344-022-10638-w. [DOI] [Google Scholar]
  283. Torres-Mendoza D, Ortega HE, Cubilla-Rios L. Patents on endophytic fungi related to secondary metabolites and biotransformation applications. J Fungi. 2020;6:58. doi: 10.3390/jof6020058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Traka MH. Health benefits of glucosinolates. Adv Bot Res. 2016;80:247–279. doi: 10.1016/bs.abr.2016.06.004. [DOI] [Google Scholar]
  285. Tyo KE, Alper HS, Stephanopoulos GN. Expanding the metabolic engineering toolbox: more options to engineer cells. Trends Biotechnol. 2007;25(3):132–137. doi: 10.1016/j.tibtech.2007.01.003. [DOI] [PubMed] [Google Scholar]
  286. Urquhart AS, Hu J, Chooi YH, Idnurm A. The fungal gene cluster for biosynthesis of the anti-bacterial agent viriditoxin. Fungal Biol Biotechnol. 2019;6:9. doi: 10.1186/s40694-019-0072-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Uzor PF, Odimegwu DC, Ebrahim W, Osadebe PO, Nwodo NJ, Okoye FB, Liu Z, Proksch P. Anti-respiratory syncytial virus compounds from two endophytic fungi isolated from Nigerian medicinal plants. Drug Res. 2016;66(10):527–531. doi: 10.1055/s-0042-111008. [DOI] [PubMed] [Google Scholar]
  288. Vahabi K, Camehl I, Sherameti I, Oelmüller R. Growth of Arabidopsis seedlings on high fungal doses of Piriformospora indica has little effect on plant performance, stress, and defense gene expression in spite of elevated jasmonic acid and jasmonic acid-isoleucine levels in the roots. Plant Signal Behav. 2013;8(11):e26301. doi: 10.4161/psb.26301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Venieraki A, Dimou M, Katinakis P. Endophytic fungi residing in medicinal plants have the ability to produce the same or similar pharmacologically active secondary metabolites as their hosts. Hell Plant Prot J. 2017;10(2):51–66. [Google Scholar]
  290. Venkateswarulu N, Shameer S, Bramhachari PV, Basha ST, Nagaraju C, Vijaya T. Isolation and characterization of plumbagin (5-hydroxyl-2-methylnaptalene-1, 4-dione) producing endophytic fungi Cladosporium delicatulum from endemic medicinal plants. Biotechnol Rep. 2018;20:e00282. doi: 10.1016/j.btre.2018.e00282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. Venugopalan A, Srivastava S. Endophytes as in vitro production platforms of high value plant secondary metabolites. Biotechnol Adv. 2015;6:873–887. doi: 10.1016/j.biotechadv.2015.07.004. [DOI] [PubMed] [Google Scholar]
  292. Verma P, Khan SA, Mathur AK, Ghosh S, Shanker K, Kalra A. Improved sanguinarine production via biotic and abiotic elicitations and precursor feeding in cell suspensions of latex-less variety of Papaver somniferum with their gene expression studies and upscaling in bioreactor. Protoplasma. 2014;251:1359–1371. doi: 10.1007/s00709-014-0638-8. [DOI] [PubMed] [Google Scholar]
  293. Verma SK, Sahu PK, Kumar K, Pal G, Gond SK, Kharwar RN, White JF. Endophyte roles in nutrient acquisition, root system architecture development and oxidative stress tolerance. J Appl Microbiol. 2021;131(5):2161–2177. doi: 10.1111/jam.15111. [DOI] [PubMed] [Google Scholar]
  294. Vignaroli C, Pasquaroli S, Citterio B, Di Cesare A, Mangiaterra G, Fattorini D, Biavasco F. Antibiotic and heavy metal resistance in enterococci from coastal marine sediment. Environ Pollut. 2018;237:406–413. doi: 10.1016/j.envpol.2018.02.073. [DOI] [PubMed] [Google Scholar]
  295. Vigneshwari A, Rakk D, Németh A, Kocsubé S, Kiss N, Csupor D, Papp T, Škrbić B, Vágvölgyi C, Szekeres A. Host metabolite producing endophytic fungi isolated from Hypericum perforatum. PLoS ONE. 2019;14(5):e0217060. doi: 10.1371/journal.pone.0217060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Vinale F, Nicoletti R, Borrelli F, Mangoni A, Parisi OA, Marra R, Lombardi N, Lacatena F, Grauso L, Finizio S, Lorito M. Co-culture of plant beneficial microbes as source of bioactive metabolites. Sci Rep. 2017;7(1):14330. doi: 10.1038/s41598-017-14569-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Vinale F, Nicoletti R, Lacatena F, Marra R, Sacco A, Lombardi N, d’Errico G, Digilio MC, Lorito M, Woo SL. Secondary metabolites from the endophytic fungus Talaromyces pinophilus. Nat Prod Res. 2017;31:1778–1785. doi: 10.1080/14786419.2017.1290624. [DOI] [PubMed] [Google Scholar]
  298. Wakefield J, Hassan HM, Jaspars M, Ebel R, Rateb ME. Dual induction of new microbial secondary metabolites by fungal bacterial co-cultivation. Front Microbiol. 2017;8:1284. doi: 10.3389/fmicb.2017.01284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Wang S, Li XM, Teuscher F, Li DL, Diesel A, Ebel R, Proksch P, Wang BG. Chaetopyranin, a benzaldehyde derivative, and other related metabolites from Chaetomium globosum, an endophytic fungus derived from the marine red alga Polysiphonia urceolata. J Nat Prod. 2006;69(11):1622–1625. doi: 10.1021/np060248n. [DOI] [PubMed] [Google Scholar]
  300. Wang FP, Chen QH, Liu XY. Diterpenoid alkaloids. Nat Prod Rep. 2010;27(4):529–570. doi: 10.1039/B916679C. [DOI] [PubMed] [Google Scholar]
  301. Wang XJ, Min CL, Ge M, Zuo RH. An endophytic sanguinarine-producing fungus from Macleaya cordata, Fusarium proliferatum BLH51. Curr Microbiol. 2014;68(3):336–341. doi: 10.1007/s00284-013-0482-7. [DOI] [PubMed] [Google Scholar]
  302. Wang X, Wu F, Liu L, Liu X, Che Y, Keller NP, Guo L, Yin WB. The bZIP transcription factor PfZipA regulates secondary metabolism and oxidative stress response in the plant endophytic fungus Pestalotiopsis fici. Fungal Genet Biol. 2015;81:221–228. doi: 10.1016/j.fgb.2015.03.010. [DOI] [PubMed] [Google Scholar]
  303. Wang X, Zhang X, Liu L, Xiang M, Wang W, Sun X, Che Y, Guo L, Liu G, Guo L, Wang C. Genomic and transcriptomic analysis of the endophytic fungus Pestalotiopsis fici reveals its lifestyle and high potential for synthesis of natural products. BMC Genomics. 2015;16(1):1–13. doi: 10.1186/s12864-014-1190-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  304. Wang Y, Li H, Feng G, Du L, Zeng D. Biodegradation of diuron by an endophytic fungus Neurospora intermedia DP8-1 isolated from sugarcane and its potential for remediating diuron-contaminated soils. PLoS ONE. 2017;12:e0182556. doi: 10.1371/journal.pone.0182556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  305. Wang Q, Cobine PA, Coleman JJ. Efficient genome editing in Fusarium oxysporum based on CRISPR/Cas9 ribonucleoprotein complexes. Fungal Genet Biol. 2018;117:21–29. doi: 10.1016/j.fgb.2018.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  306. Wang H, Zhang R, Duan Y, Jiang W, Chen X, Shen X, Yin C, Mao Z. The endophytic strain Trichoderma asperellum 6S–2: an efficient biocontrol agent against apple replant disease in China and a potential plant-growth-promoting fungus. J Fungi. 2021;7(12):1050. doi: 10.3390/jof7121050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Wani ZA, Mirza DN, Arora P, Riyaz-Ul-Hassan S. Molecular phylogeny, diversity, community structure, and plant growth-promoting properties of fungal endophytes associated with the corms of saffron plant: an insight into the microbiome of Crocus sativus Linn. Fungal Biol. 2016;120(12):1509–1524. doi: 10.1016/j.funbio.2016.07.011. [DOI] [PubMed] [Google Scholar]
  308. Wei Q, Bai J, Yan D, Bao X, Li W, Liu B, Zhang D, Qi X, Yu D, Hu Y. Genome mining combined metabolic shunting and OSMAC strategy of an endophytic fungus leads to the production of diverse natural products. Acta Pharm Sin B. 2021;11(2):572–587. doi: 10.1016/j.apsb.2020.07.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Wen Y, Lv Y, Hao J, Chen H, Huang Y, Liu C, Huang H, Ma Y, Yang X. Two new compounds of Penicillium polonicum, an endophytic fungus from Camptotheca acuminate Decne. Nat Prod Res. 2020;34:1879–1883. doi: 10.1080/14786419.2019.1569003. [DOI] [PubMed] [Google Scholar]
  310. Widjajanti H, Nurnawati E, Tripuspita V. The potency of endophytic fungi isolated from Hippobroma longiflora (L) G. Don as an anti-oxidant sources. IOP Conf Ser Earth Environ Sci. 2022;976:012045. doi: 10.1088/1755-1315/976/1/012045. [DOI] [Google Scholar]
  311. Wu W, Tran W, Taatjes CA, Alonsogutierrez J, Lee TS, Gladden JM. Rapid discovery and functional characterization of terpene synthases from four endophytic Xylariaceae. PLoS ONE. 2016;11(2):e0146983. doi: 10.1371/journal.pone.0146983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Wu W, Davis RW, Tran-Gyamfi MB, Kuo A, Labutti K, Mihaltcheva S, Hundley H, Chovatia M, Lindquist E, Barry K. Characterization of four endophytic fungi as potential consolidated bioprocessing hosts for conversion of lignocellulose into advanced biofuels. Appl Microbiol Biotechnol. 2017;101(6):2603–2618. doi: 10.1007/s00253-017-8091-1. [DOI] [PubMed] [Google Scholar]
  313. Wu JS, Shi XH, Zhang YH, Yu JY, Fu XM, Li X, Chen KX, Guo YW, Shao CL, Wang CY. Co-cultivation with 5-azacytidine induced new metabolites from the zoanthid-derived fungus Cochliobolus lunatus. Front Chem. 2019;7:763. doi: 10.3389/fchem.2019.00763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Xia Y, Sahib MR, Amna A, Opiyo SO, Zhao Z, Gao YG. Culturable endophytic fungal communities associated with plants in organic and conventional farming systems and their effects on plant growth. Sci Rep. 2019;9(1):1–10. doi: 10.1038/s41598-018-38230-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Xiao X, Luo S, Zeng G, Wei W, Wan Y, Chen L, Guo H, Cao Z, Yang L, Chen J. Biosorption of cadmium by endophytic fungus (EF) Microsphaeropsis sp. LSE10 isolated from cadmium hyperaccumulator Solanum nigrum L. Bioresour Technol. 2010;101(6):1668–1674. doi: 10.1016/j.biortech.2009.09.083. [DOI] [PubMed] [Google Scholar]
  316. Xie XG, Fu WQ, Zhang FM, Shi XM, Zeng YT, Li H, Zhang W, Dai CC. The endophytic fungus Phomopsis liquidambari increases nodulation and N2 fixation in Arachis hypogaea by enhancing hydrogen peroxide and nitric oxide signaling. Microb Ecol. 2017;74(2):427–440. doi: 10.1007/s00248-017-0944-8. [DOI] [PubMed] [Google Scholar]
  317. Xiong ZQ, Yang YY, Na Z, Yong W. Diversity of endophytic fungi and screening of fungal paclitaxel producer from Anglojap yew, Taxus x Media. BMC Microbiol. 2013;13(1):71. doi: 10.1186/1471-2180-13-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Xu J, Kjer J, Sendker J, Wray V, Guan H, Edrada R, Lin W, Wu J, Proksch P. Chromones from the endophytic fungus Pestalotiopsis sp. isolated from the Chinese mangrove plant Rhizophora mucronata. J Nat Prod. 2009;72(4):662–665. doi: 10.1021/np800748u. [DOI] [PubMed] [Google Scholar]
  319. Xu G, Yang S, Meng L, Wang BG. The plant hormone abscisic acid regulates the growth and metabolism of endophytic fungus Aspergillus nidulans. Sci Rep. 2018;8(1):1–9. doi: 10.1038/s41598-018-24770-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. Xu G, Yang S, Meng L, Wang BG. The plant hormone abscisic acid regulates the growth and metabolism of endophytic fungus Aspergillus nidulans. Sci Rep. 2018;8:1–9. doi: 10.1038/s41598-018-24770-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Xue M, Hou X, Fu J, Zhang J, Wang J, Zhao Z, Xu D, Lai D, Zhou L. Recent advances in search of bioactive secondary metabolites from fungi triggered by chemical epigenetic modifiers. J Fungi. 2023;9(2):172. doi: 10.3390/jof9020172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Yadav AN. Biodiversity and biotechnological applications of host-specific endophytic fungi for sustainable agriculture and allied sectors. Acta Sci Microbiol. 2018;1(5):1–5. [Google Scholar]
  323. Yadav R, Singh AV, Kumar M, Yadav S. Phytochemical analysis and plant growth promoting properties of endophytic fungi isolated from Tulsi and Aloe vera. Int J Agric Stat Sci. 2016;12(1):239–248. [Google Scholar]
  324. Yan Z, Shi J, Gao Z, Yangwu R, Jiang H, Che J, Liu Y. Production of pinoresinol diglucoside, pinoresinol monoglucoside, and pinoresinol by Phomopsis sp. XP-8 using mung bean and its major components. Appl Microbiol Biotechnol. 2015;99(11):4629–4643. doi: 10.1007/s00253-015-6491-7. [DOI] [PubMed] [Google Scholar]
  325. Yan L, Zhao H, Zhao X, Xu X, Di Y, Jiang C, Shi J, Shao D, Huang Q, Yang H, Jin M. Production of bioproducts by endophytic fungi: chemical ecology, biotechnological applications, bottlenecks, and solutions. Appl Microbiol Biotechnol. 2018;102(15):6279–6298. doi: 10.1007/s00253-018-9101-7. [DOI] [PubMed] [Google Scholar]
  326. Yan L, Zhao H, Zhao X, Xu X, Di Y, Jiang C, Shi J, Shao D, Huang Q, Yang H, Jin M. Production of bioproducts by endophytic fungi: chemical ecology, biotechnological applications, bottlenecks, and solutions. Appl Microbiol Biotechnol. 2019;102(15):6279–6298. doi: 10.1007/s00253-018-9101-7. [DOI] [PubMed] [Google Scholar]
  327. Yang Y, Cheng X, Liu W, Chou G, Wang Z, Wang C. Potent AChE and BChE inhibitors isolated from seeds of Peganum harmala Linn by a bioassay-guided fractionation. J Ethnopharmacol. 2015;168:279–286. doi: 10.1016/j.jep.2015.03.070. [DOI] [PubMed] [Google Scholar]
  328. Yang ZD, Li ZJ, Zhao JW, Sun JH, Yang LJ, Shu ZM. Secondary metabolites and PI3K inhibitory activity of Colletotrichum gloeosporioides, a fungal endophyte of Uncaria rhynchophylla. Curr Microbiol. 2019;76:904–908. doi: 10.1007/s00284-019-01707-7. [DOI] [PubMed] [Google Scholar]
  329. Yang W, Chen X, Li Y, Guo S, Wang Z, Yu X. Advances in pharmacological activities of terpenoids. Nat Prod Commun. 2020;15(3):1934578X20903555. doi: 10.1177/1934578X20903555. [DOI] [Google Scholar]
  330. Yang Z, Wu K, Xu Y, Xia X, Wang X, Ge M, Shao L. Three novel chromanones with biological activities from the endophytic fungus Phomopsis CGMCC No. 5416. J Antibiot Res. 2020;73:194–199. doi: 10.1038/s41429-019-0270-0. [DOI] [PubMed] [Google Scholar]
  331. Yang L, Zou YN, Tian ZH, Wu QS, Kuča K. Effects of beneficial endophytic fungal inoculants on plant growth and nutrient absorption of trifoliate orange seedlings. Sci Hortic. 2021;277:109815. doi: 10.1016/j.scienta.2020.109815. [DOI] [Google Scholar]
  332. Yao YR, Tian XL, Shen BM, Mao ZC, Chen GH, Xie BY. Transformation of the endophytic fungus Acremonium implicatum with GFP and evaluation of its biocontrol effect against Meloidogyne incognita. World J Microbiol Biotechnol. 2015;31(4):549–556. doi: 10.1007/s11274-014-1781-2. [DOI] [PubMed] [Google Scholar]
  333. Yao YQ, Lan F, Qiao YM, Wei JG, Huang RS, Li LB. Endophytic fungi harbored in the root of Sophora tonkinensis Gapnep: diversity and biocontrol potential against phytopathogens. Microbiologyopen. 2017;6(3):e00437. doi: 10.1002/mbo3.437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  334. Yedukondalu N, Arora P, Wadhwa B, Malik FA, Vishwakarma RA, Gupta VK, Riyaz-Ul-Hassan S, Ali A. Diapolic acid A-B from an endophytic fungus, Diaporthe terebinthifolii depicting anti-microbial and cytotoxic activity. J Antibiot. 2017;70(2):212–215. doi: 10.1038/ja.2016.109. [DOI] [PubMed] [Google Scholar]
  335. You X, Feng S, Luo S, Cong D, Yu Z, Yang Z, Zhang J. Studies on a rhein-producing endophytic fungus isolated from Rheum palmatum L. Fitoterapia. 2013;85(1):161–168. doi: 10.1016/j.fitote.2012.12.010. [DOI] [PubMed] [Google Scholar]
  336. Yu QY, Fang L, Yun MQ, Ji GW, Rong SH, Liang BL. Endophytic fungi harbored in the root of Sophora tonkinensis Gapnep: diversity and biocontrol potential against phytopathogens. Microbiologyopen. 2017;6(3):e437. doi: 10.1002/mbo3.437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Yuan J, Sun K, Deng-Wang MY, Dai CC. The mechanism of ethylene signaling induced by endophytic fungus Gilmaniella sp. AL12 mediating sesquiterpenoids biosynthesis in Atractylodes lancea. Front Plant Sci. 2016;7:361. doi: 10.3389/fpls.2016.00361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  338. Yuan Y, Cheng S, Bian G, Yan P, Ma Z, Dai W, Chen R, Fu S, Huang H, Chi H, Cai Y. Efficient exploration of terpenoid biosynthetic gene clusters in filamentous fungi. Nat Catal. 2022;5(4):277–287. doi: 10.1038/s41929-022-00762-x. [DOI] [Google Scholar]
  339. Zahoor M, Irshad M, Rahman H, Qasim M, Afridi SG, Qadir M, Hussain A. Alleviation of heavy metal toxicity and phytostimulation of Brassica campestris L. by endophytic Mucor sp. MHR-7. Ecotoxicol Environ Safe. 2017;142:139–149. doi: 10.1016/j.ecoenv.2017.04.005. [DOI] [PubMed] [Google Scholar]
  340. Zazopoulos E, Huang K, Staffa A, Liu W, Bachmann BO, Nonaka K, Ahlert J, Thorson JS, Shen B, Farnet CMA. Genomics-guided approach for discovering and expressing cryptic metabolic pathways. Nat Biotech. 2003;21(2):187–190. doi: 10.1038/nbt784. [DOI] [PubMed] [Google Scholar]
  341. Zhai X, Luo D, Li X, Han T, Jia M, Kong Z, Ji J, Rahman K, Qin L, Zheng C. Endophyte Chaetomium globosum D38 promotes bioactive constituents accumulation and root production in Salvia miltiorrhiza. Front Microbiol. 2018;8:2694. doi: 10.3389/fmicb.2017.02694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  342. Zhan J, Burns AM, Liu MX, Faeth SH, Gunatilaka AL. Search for cell motility and angiogenesis inhibitors with potential anti-cancer activity: beauvericin and other constituents of two endophytic strains of Fusarium oxysporum. J Nat Prod. 2007;70(2):227–232. doi: 10.1021/np060394t. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Zhang P, Zhou PP, Yu LJ. An endophytic taxol-producing fungus from Taxus x media, Aspergillus candidus MD3. FEMS Microbiol Lett. 2009;293(2):155–159. doi: 10.1111/j.1574-6968.2009.01481.x. [DOI] [PubMed] [Google Scholar]
  344. Zhang Y, Jia A, Chen H, Wang M, Ding G, Sun L, Li L, Dai M. Anthraquinones from the saline–alkali plant endophytic fungus Eurotium rubrum. J Antibiot Res. 2017;70:1138–1141. doi: 10.1038/ja.2017.121. [DOI] [PubMed] [Google Scholar]
  345. Zheng YST, Qi Q. Microbial CRISPRi and CRISPRa systems for metabolic engineering. Biotechnol Bioprocess Eng. 2019;24:579–591. doi: 10.1007/s12257-019-0107-5. [DOI] [Google Scholar]
  346. Zhi-lin Y, Chuan-chao D, Lian-qing C. Regulation and accumulation of secondary metabolites in plant-fungus symbiotic system. Afric J Biotech. 2007;6(11):1266–1271. [Google Scholar]
  347. Zhu M, Zhang X, Feng H, Che Q, Zhu T, Gu Q, Li D. Campyridones A-D, pyridone alkaloids from a mangrove endophytic fungus Campylocarpon sp. HDN13–307. Tetrahedron. 2016;72:5679–5683. doi: 10.1016/j.tet.2016.07.080. [DOI] [Google Scholar]
  348. Zhu Y, Shao Y, Li L, Zhao L, Zhang M, Dong C. The plant growth-promoting endophytic Fusarium oxysporum GG22 enhances Rehmannia glutinosa secondary metabolites accumulation. Ind Crops Prod. 2022;182:114881. doi: 10.1016/j.indcrop.2022.114881. [DOI] [Google Scholar]
  349. Zou R, Wei C, Zhang X, Zhou D, Xu J. Alkaloids from endophytic fungus Aspergillus fumigatus HQD24 isolated from the Chinese mangrove plant Rhizophora mucronata. Nat Prod Res. 2021 doi: 10.1080/14786419.2021.1916017. [DOI] [PubMed] [Google Scholar]
  350. Zouiten A, Beltifa A, Van Loco J, Mansour HB, Reyns T. Ecotoxicological potential of antibiotic pollution–industrial wastewater: bioavailability, biomarkers, and occurrence in Mytilus galloprovincialis. Environ Sci Pollut Res. 2016;23(15):15343–15350. doi: 10.1007/s11356-016-6713-2. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from 3 Biotech are provided here courtesy of Springer

RESOURCES