Next Article in Journal
Altered TRPM7-Dependent Calcium Influx in Natural Killer Cells of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Patients
Next Article in Special Issue
Pharmacological Potential of Betulin as a Multitarget Compound
Previous Article in Journal
Targeted Disruption of the MORG1 Gene in Mice Causes Embryonic Resorption in Early Phase of Development
Previous Article in Special Issue
The Outstanding Chemodiversity of Marine-Derived Talaromyces
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fungal Endophytes: Microfactories of Novel Bioactive Compounds with Therapeutic Interventions; A Comprehensive Review on the Biotechnological Developments in the Field of Fungal Endophytic Biology over the Last Decade

1
School of Studies in Biotechnology, Pandit Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
2
Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana 133207, Haryana, India
3
Department of Pharmaceutics, Unaizah College of Pharmacy, Qassim University, Unaizah 51911, Saudi Arabia
4
Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2023, 13(7), 1038; https://doi.org/10.3390/biom13071038
Submission received: 8 May 2023 / Revised: 14 June 2023 / Accepted: 15 June 2023 / Published: 25 June 2023
(This article belongs to the Special Issue New Advances in Natural Products in Drug Discovery)

Abstract

:
The seminal discovery of paclitaxel from endophytic fungus Taxomyces andreanae was a milestone in recognizing the immense potential of endophytic fungi as prolific producers of bioactive secondary metabolites of use in medicine, agriculture, and food industries. Following the discovery of paclitaxel, the research community has intensified efforts to harness endophytic fungi as putative producers of lead molecules with anticancer, anti-inflammatory, antimicrobial, antioxidant, cardio-protective, and immunomodulatory properties. Endophytic fungi have been a valuable source of bioactive compounds over the last three decades. Compounds such as taxol, podophyllotoxin, huperzine, camptothecin, and resveratrol have been effectively isolated and characterized after extraction from endophytic fungi. These findings have expanded the applications of endophytic fungi in medicine and related fields. In the present review, we systematically compile and analyze several important compounds derived from endophytic fungi, encompassing the period from 2011 to 2022. Our systematic approach focuses on elucidating the origins of endophytic fungi, exploring the structural diversity and biological activities exhibited by these compounds, and giving special emphasis to the pharmacological activities and mechanism of action of certain compounds. We highlight the tremendous potential of endophytic fungi as alternate sources of bioactive metabolites, with implications for combating major global diseases. This underscores the significant role that fungi can play in the discovery and development of novel therapeutic agents that address the challenges posed by prevalent diseases worldwide.

1. Introduction

Prominent amongst modern day healthcare challenges is the emergence of resistance among pathogenic microorganisms, novel occurrences of life-threatening viruses, and rising incidences of communicable and noncommunicable diseases. These medical challenges provide an urgent and compelling need to harness and leverage novel resources that offer sustainable solutions [1,2,3]. Natural products, which are metabolites or by-products derived from plants, animals, or microorganisms, have always been used to treat various kinds of human ailments. The novel structures and frameworks of these compounds, combined with their broad-spectrum activities and potential as lead molecules, offer immense promise in therapeutic applications [4,5]. Natural products have the potential to be directly used as drugs or as building blocks for the synthesis of new drugs through combinatorial synthesis methods. They can either be utilized in their original form or modified to synthesize novel compounds with enhanced pharmacological properties. It is noteworthy that approximately 55% of the drugs that have been approved for clinical use in the last three decades can be traced back to natural products, whereas 58% of drugs have been developed by imitating natural product structures [6,7,8]. For centuries, plants have served as a key source of phytochemicals for drug discovery and development [5,9]. The serendipitous discovery of penicillin in 1928 by Sir Alexander Fleming from Penicillium chrysogenum marked the beginning of the golden era of antibiotics. The subsequent success of several lifesaving drugs obtained from microorganisms, such as the cholesterol biosynthesis inhibitor lovastatin from Aspergillus terrus and the immunosuppressant cyclosporine from Tolypocladium inflatum has brought about a significant change in drug discovery and development, shifting the focus from plants to microorganisms [1,2,10]. Since then, fungi have played a significant role in benefiting human welfare through the production of bioactive compounds that have been utilized as antimicrobial [3,4], anticancer [11,12], antioxidant [13], and immunomodulatory agents [14]. However, even after the pioneering discovery of penicillin ninety-five years ago, fungi continue to be the most underexplored biosource of natural products, particularly considering their vast biodiversity, unique biochemical properties, and significant biotechnological potential. This is despite the ongoing characterization of over a thousand fungal species annually, with several thousand more awaiting isolation and further characterization. As a result, the utility of fungal products largely remains unexplored and untapped, despite the impressive new taxonomic findings. In addition, the complexity of fungal biosynthetic pathways, as revealed by whole genome sequencing and subsequent genome mining of various fungal species, poses further challenges to harnessing the full potential of fungi [5,15,16,17,18].
To overcome the limitations faced by existing methods in the field of fungal bioprospecting, it is essential to adopt novel screening strategies that can effectively identify fungi inhabiting distinct ecological environments. One potential strategy involves targeting fungi that establish mutualistic alliances with plants, residing within their living tissues without causing any apparent symptoms. This particular group of fungi, known as endophytes, holds great promise as a source of bioactive compounds [3,16,19,20]. Endophytic fungi secrete an array of bioactive compounds that serve multiple functions, such as stimulating plant growth, inducing defense mechanisms against pathogens, and serving as agents for remediating salt and drought stresses [3,21,22,23,24]. This coevolution between endophytic fungi and their host plants results in the production of bioactive compounds which contribute in a variety of ways to plant–microbe interactions and can provide fitness benefits to the host plant (Figure 1) [25,26,27,28]. Endophytic fungi establish their communication with their host plants through metabolic interactions [29,30]. According to the xenohormesis hypothesis [31], heterotrophic organisms such as fungi, under selective evolutionary pressure, develop the ability to sense stress-induced chemical cues from host plants and start producing analogous chemicals themselves. In essence they mimic the biological properties of the host plant [27,32]. In addition to synthesizing compounds that are analogous to host plant compounds, endophytic fungi also exhibit a vast repertoire of diverse secondary metabolites with intriguing biological and/or pharmaceutical properties. In the last thirty years, a wide range of bioactive compounds with potential in healthcare and medicine have been discovered from endophytic fungi. These compounds exhibit various properties such as antimicrobial [1,3,18], anticancer [33,34], antioxidant [35,36,37,38], anti-inflammatory [39,40], antidiabetic [41,42,43], and immunosuppressive activities [44,45,46,47]. The abundance of such biologically active metabolites derived from endophytic fungi highlights their importance as a valuable source of potential therapeutic substances [3,4,5,18,22,34]

2. What Is an Endophyte?

There exists a vast number of plant species on earth, exceeding 300,000, and each of these plants hosts a diverse range of microorganisms, broadly categorized as either epiphytes, endophytes, or pathogens [3,48]. Among them, endophytes comprise a diverse group of ubiquitous, polyphyletic microorganisms that reside within plant cells or in the intracellular space for at least a part of their life cycle, without showing any external manifestation of their presence [22,49,50,51,52]. Fossil records indicate that the microorganisms associated with plants can be traced back over 400 million years to the Devonian period, suggesting that the alliance between plants and endophytes may have originated during the early emergence of land plants on earth [27,30,53]. The term “endophyte” is derived from its literal meaning of “within the plants” (“endon” meaning within; “phyton” meaning plants) [54]. In 1866, the German botanist Anton de Bary coined the term “endophytes” to describe organisms that live within plants without any visible symptoms; however, the first endophytes were discovered in 1904 from a Eurasian darnel ryegrass, Lolium temulentum [23,55,56]. Endophytes can be found thriving in a range of ecological niches including the Artic and the Antarctic regions, deserts, mangroves, rainforests, as well as marine and coastal ecosystems [9,24,51,57,58]. Endophytes exhibit diverse relationships with their host plants including symbiotic, benign commensal, decomposer, and latent pathogenic interactions [3,59]. Once an endophyte successfully colonizes the internal tissue of a host plant, it enters a dormant phase that can persist for its entire lifecycle or for an extended duration until favorable conditions arise. During this period, endophytes remain inactive or exhibit minimal metabolic activity, waiting for environmental cues that indicate the availability of suitable conditions for growth and proliferation. This coevolutionary process creates a mutually beneficial relationship between the host plant and the endophyte. The host plant supplies vital nutrients and shelter to the endophyte that are required for its survival, while the endophyte reciprocates by producing bioactive metabolites that enhance the fitness of the host plant. The bioactive metabolites produced by endophytes play a crucial role in enabling host plants to withstand biotic and abiotic stresses, conserve water, and defend themselves against microbial, pest, and insect attack. As a result, endophytes play a vital role in plant symbiosis by providing protection to their host plants against pathogenic threats and challenging environmental conditions [9,24,25,32,60]. Endophytes continuously adapt and evolve in response to biotic and abiotic stresses forming intricate interactions (bi-, tri-, or multipartite) with their host plant. This symbiotic relationship leads to the production of valuable natural products with therapeutic potential. These bioactive compounds, produced through the ongoing process of the strain development of endophytes, can be utilized directly or indirectly as therapeutic agents. The dynamic interplay between endophytes and their host plants gives rise to a diverse range of bioactive metabolites that holds promise for various therapeutic applications [17,25,61]. Through genetic recombination with the host plant, endophytes also acquire the ability to emulate the biological properties of their host plant and produce analogous bioactive metabolites. This proficiency in metabolism makes them a highly valuable resource for the exploration and discovery of natural bioactive metabolites [5,17,25].

3. Exploring Bioactive Metabolites from Endophytic Fungi: Unveiling Nature’s Treasure Trove

Endophytes, despite being isolated as early as 1904, remained largely overlooked for a considerable period of time. Apart from sporadic research, the biochemical research community did not pay much attention to endophytes until the discovery of Taxomyces andreanae, an endophytic fungus isolated from Pacific yew (Taxus brevifolia) in 1993. This fungus demonstrated an extraordinary capability to independently produce the highly successful anticancer drug taxol in its culture broth, resembling its host [62,63]. This breakthrough discovery initiated a global quest among researchers to delve deeper into the exploration of endophytic fungi with the aim of uncovering potential bioactive compounds [20,25,30,32,51,57,58,62,64,65]. Following this, significant findings emerged which unveiled the potential of endophytes to synthesize analogous bioactive metabolites with notable therapeutic properties. Compounds such as taxol, resveratrol, huperzine, camptothecin, podophyllotoxin, and vinca alkaloids were among those discovered from endophytic fungi, showcasing their ability to produce bioactive compounds that can be used as therapeutic agents for the treatment of diverse ailments, either through direct application or indirect utilization (Table 1, Figure 2) [23,25,52,66,67,68,69]. Furthermore, endophytic fungi are also capable of producing a wide range of nonanalogous compounds that exhibit significant bioactivities. The bioactive metabolites derived from endophytes predominantly belong to the chemical class of alkaloids, cytochalasins, flavonoids, polyketides, steroids, and terpenoids [70,71]. The metabolites produced by endophytes have been found to display a wide range of pharmacological properties primarily encompassing antimicrobial, antineoplastic, anticancer, antioxidant, anti-inflammatory, antidiabetic, and antidepressant activities [2,3,4,22,25,34,52,72]. In addition, endophytes have been identified as a viable source of numerous enzymes such as amylase, catalase, laccase, lipase, and proteases that have significant clinical and industrial applications [40,58,73]. Thus, endophytic microorganisms represent a valuable reservoir of bioactive secondary metabolites with tremendous potential in the agrochemical and pharmaceutical industries [2,8,9,51,74,75]. This review highlights significant bioactive molecules discovered from endophytic fungi over the last decade, along with their potential applications in the treatment of various life-threatening diseases. The article presents a comprehensive analysis of 296 newly discovered compounds derived from endophytic fungi, characterized by novel or rare structures or skeletal frameworks across 290 journal articles published between 2011 and 2022. Furthermore, the article provides a concise overview of the origin of these endophytic fungi, the chemical structures of the compounds, and their corresponding biological activities.

3.1. Anticancer Compounds from Endophytic Fungi

Cancer is a complex and diverse group of diseases characterized by uncontrolled growth and spread of abnormal cells in the body. It is a major contributor to global mortality and the future outlook indicates an upward trend in cancer-related deaths. The World Health Organization (WHO) has estimated that around ten million people died from cancer worldwide in 2020, and this figure is anticipated to reach 13.1 million by the year 2030 [95]. The most prevalent types of cancer globally include breast, cervical, colon, prostrate, oral, rectal, skin, and stomach cancer. Cancer incidences exhibit significant variation across countries and regions with higher rates observed in more developed nations. The incidence of cancer is influenced by several factors, including genetics, lifestyle choices, and access to healthcare [96]. The primary treatment options for the aforementioned types of cancer include surgery, radiation, immuno- and chemotherapy. While these treatments can be effective to a certain extent, they often come with significant side effects such as weakness, hair loss, cognitive issues, and increased vulnerability to infections. Additionally, some cancer cells develop resistance to these drugs, which diminishes the effectiveness of these therapies [97]. To address these challenges, ongoing research is dedicated to developing novel anticancer compounds and therapies that offer a precise targeting of cancer cells and have fewer side effects [98]. In recent years, there has been a considerable interest in fungal endophytes as a potential source of new drugs. This interest stems from the remarkable discovery of the anticancer drug “taxol” in the endophytic fungus T. andreanae isolated from the Pacific yew tree [62,99]. This breakthrough instigated a widespread initiative to systematically screen diverse plant species for the presence of taxol-producing endophytes. This approach has been successful in finding taxol-producing endophytes not only in the taxus plant but also in various other plant species. Extensive studies have identified taxol or its analogue-producing endophyte in various fungal genera, including Alternaria, Bartalinia, Fusarium, Lasiodiplodia, Metarhizium, Monochaetia, Pestalotiopsis, Penicillium, Phoma, Pithomyces, Seimatoantlerium, Sporormia, Trichothecium, Tubercularia, and Truncatella. Originally, taxol was found to be active against L-1210, P-388, and P-1534 leukemias whereas now, taxol is primarily used in combination with other anticancer drugs for the treatment of breast, ovarian, lung, and advanced testicular cancers [23,100].
Vinblastine and vincristine (also known as vinca alkaloids) are plant-based chemotherapeutic agents that exhibit therapeutic activity by binding to microtubule and spindle proteins, leading to cell-cycle arrest and apoptotic cell death in cancer cells. Initially isolated from Madagascar periwinkle plant (Catharanthus roseus), these vinca alkaloids have been widely employed in the treatment of various cancer types. The discovery of vinblastine and vincristine sparked a global quest to explore alternative sources of these valuable compounds [101,102]. Fungal endophytes such as Eutypella sp., Fusarium oxysporum, Nigrospora sphaerica, and Talaromyces radicus isolated from Madagascar periwinkle plant have been discovered to produce vinblastine and vincristine. These compounds have exhibited cytotoxic activity in a dose-dependent manner against HeLa, MCF-7, A-549, U-251, A-431, and MDA-MB 231 cancer cell lines [79,80,82,83].
Camptothecin, a pentacyclic quinolone alkaloid, is primarily sourced from the wood of Camptotheca acuminate (a Chinese ornamental plant) and the roots of Nothapodytes foetida [67,87,103]. Camptothecin is the third largest plant-based antineoplastic agent that executes its cytotoxic property by selectively inhibiting topoisomerase I, an enzyme which plays a vital role in DNA replication [23,102]. In recent years, several fungal endophytes such as Aspergillus sp. LY341, LY355, Alternaria burnsi, F. solani S-019, and Trichoderma atroviride LY357 have been found to produce camptothecin, which has a cytotoxic effect on human breast, lung, and ovarian cancer cell lines [86,87,104].
Podophyllotoxin is a highly valued aryltetralin lignin that serves as a precursor for the synthesis of anticancer drugs including etoposide, teniposide, and etopophos phosphate, which are clinically used for treating bronchial and testicular cancers [23]. Podophyllotoxin has a potent inhibitory effect on microtubule assembly, while its derivatives, etoposide and teniposide inhibit the activity of the topoisomerase enzyme II, resulting in cell-cycle arrest in the S phase [89]. Notably, certain endophytic fungi including Mucor fragilis TW5 and A. tenuissima have been identified as producers of podophyllotoxin, which exhibits cytotoxic activity against human colon, lung, and prostate cancer cell lines [89,105].
Potent cytotoxic activity has been observed in endophytic fungi such as Pestalotiopsis palmarum, Pestalotiopsis sp. FT172, and P. uvicola, isolated from the Chinese medicinal plants Sinomenium acutum, Myrsine sandwicensis, and Artemisia japonica, respectively. These endophytes secrete bioactive compounds such as ambuic acid, genistein, kaempferol, quercetin, and rutin that have demonstrated cytotoxic activity against HeLa, HCT116, A549, A2780, and drug-resistant breast, ovarian, and cisplatin-resistant A2789 (A2780CisR) cancer cell lines. Similarly, novel compounds including phomopchalasin B and C derived from Phomopsis sp. sh2 as well as mycoepoxydiene, deacetylmycoepoxydiene, phomoxydiene C, and cytosporone E obtained from Phomopsis sp. BCC 45011 exhibited cytotoxic activity against HL-60, SMMC-7221, A-549 KB, MCF-7, NCI-H187, and vero cell lines, respectively [34].
Xylaria psidii, an endophytic fungus isolated from the leaves of Aegle marmelos, yielded two notable compounds, xylarione A and (−) 5-methylmellein, which exhibited cytotoxic activity against MCF-7, MIA-Pa-Ca-2, NCI-H226, HepG2, and DU-145 cancer cell lines with an IC50 value ranging from 16 to 37 µM [106]. Similarly, cytochalasin Q, a bioactive compound isolated from endophytic Xylaria sp. ZJWCF255 displayed cytotoxic activity against SMMC-772, MCF-7, and MGC80-3 cancer cell lines [34]. Furthermore, endophytic Chaetomium globosum isolated from Ginko biloba produced chaetoglobosin A, which showed remarkable cytotoxicity against HCT-116 cell lines with an IC50 values in the range of 3.15–8.44 µM [107]. The identification of endophytic fungi as a lucrative source of anticancer drugs has opened up new possibilities for drug development. These fungi have been found to possess a wide range of bioactive compounds that hold potential in the fight against cancer. However, it is crucial to emphasize here that the production and development of these compounds are still in the early stages of investigation. Further studies are needed to elucidate their precise mechanisms of action, evaluate their safety profiles, and assess their suitability for clinical use. Further research is necessary to unlock the full potential of endophytic fungi as a viable source of effective and safer anticancer drugs (Table 2; Figure 3).

3.2. Antioxidant Compounds from Endophytic Fungi

Free radicals are unstable molecules that are either produced naturally in the body as a byproduct of metabolism or can be formed by external factors such as UV light, pesticides, drugs, smoking, and alcohol. These free radicals can damage cells and lead to various diseases such as diabetes, Down’s syndrome, degenerative disease, Alzheimer’s disease, Parkinson’s disease, and cardiovascular disorders [129,130,131]. Antioxidant compounds protect cellular damage by neutralizing these free radicals and preventing them from causing cellular damage. Antioxidant compounds also play a significant role in preventing cancer, as they can react with and neutralize the free radicals that contribute to the formation of cancer cells. Thus, it is important to develop new antioxidant compounds, as they can help to stabilize free radicals and prevent cellular damage, thus improving human health and preventing degenerative and other diseases [132]. Studies have shown that endophytic fungi can produce a wide variety of compounds with strong antioxidant activities, and some of them have been isolated and characterized, such as phenolic acids, xanthones, flavonoids, terpenoids, and polyketides [133]. The bioprospection of endophytic fungi is a promising area, and new antioxidant compounds from fungal endophytes are continuously being discovered and characterized. Anofinic acid obtained from endophytic A. tubenginses ASH4 showed potential antioxidant and anticancer activities [134]. Endophytic Aspergillus sp. MFLUCC16-0603, MFLUCC16-0614, and Nigrospora sp. MFLUCC16-0605 isolated from Ocimum basilicum exhibited antioxidant activity with IC50 values ranging between 11.75 and 17.39 mg/mL, respectively [135]. Similarly, endophytic A. alternata and P. citrinum isolated from Azadirachta indica have been found to have potential antioxidant activity, with IC50 values of 38–52.13 μg/mL, respectively [136]. Other endophytic species, such as Chaetomium sp., Colletotrichum sp., Curvularia sp., and Trichoderma sp., isolated from similar host plants, have also exhibited antioxidant activity ranging from 31 to 69% [137]. Five endophytic fungal isolates PAL 01-B2, PAL 01-D2, PAL 04-R2, PAL 11-B1, and PAL 14-D3 possessed strong antioxidant activities with IC50 value ranging between 5.26 and 14.06 µg/mL, respectively [138]. Potential antioxidant properties were also demonstrated by endophytic Aspergillus sp., Alternaria sp. (ML4), Chaetomium sp., Penicillium sp., and Phomopsis sp. GJJM07 isolated from Calotropis procera, Eugenia jambolana, Mesua ferrea, Trigonella foenum-graecum, and Triticum durum, respectively [139,140,141,142]. Fermentation extracts of fungal endophytes ZA 163, MO 211, LO 261, FE 082, and FE 084 associated with Nigerian ethnomedicinal plants Albizia zygia, Millettia thonningii, Alchornea cordifolia, and Ficus exasperate were found to produce pyrogallol, dl-alpha-tocopherol, Alpha tocospiro, linoleic acid, 9-octadecenamide, lupeol, and 9-octadecenoic acid (Z), which exhibited antioxidant activity [143]. Similarly, the fungal extracts of Fusarium SaR-2 and Alternaria SaF-2 have significant antioxidant properties with 90.14% and 83.25% free-radical scavenging activity, respectively [144]. Furthermore, extracts of Chaetomium globosum associated with Adiantum capillus showed 99% free-radical scavenging activity at a concentration of 100 µg/mL [145] (Table 3) (Figure 4).

3.3. Anti-Inflammatory Compounds from Endophytic Fungi

Inflammation is a multifaceted aspect of the immune response that arises in response to various factors such as pathogens, cellular injury, toxins, and radiation. It can manifest as either a short-term immediate reaction or as a long-lasting persistent condition. Inflammation has the potential to impact a wide range of body organs including the heart, pancreas, liver, kidney, lungs, brain, intestinal tract, and reproductive system. The underlying cause of inflammation can either be infectious or noninfectious and if left unresolved, it can result in tissue damage or contribute to disease development, depending upon the causative agent involved [154,155]. Studies have shown that the metabolome of endophytic fungi includes anti-inflammatory compounds similar to their host and are thus believed to be a potential source of agents for combating inflammation and improving human health. Lasiodiplactone A, derived from the marine mangrove plant Acanthus ilicifolius and produced by the endophytic fungus Lasiodiplodia theobromae demonstrated significant anti-inflammatory activity by inhibiting the production of nitric oxide (NO) in RAW 264.7 cells stimulated with lipopolysaccharide with an IC50 of 23.5 µM. In addition, it also exhibited inhibitory activity against α-glucosidase with an IC50 value of 29.4 µM [154]. Similarly, Botryosphaerin B, derived from the endophytic fungus Botryosphaeria sp. SCSIO KcF6 in the mangrove plant Kandelia candel showed an inhibitory effect on cyclooxygenase (COX)–2 activity with a significant IC50 value of 1.12 mM [155]. Cyclonerodial B obtained from the endophytic fungus Trichoderma sp. Xy24 isolated from Xylocarpus granatum exhibited anti-inflammatory properties by suppressing the production of nitric oxide (NO) in BV2 microglia cells. This compound also has potential therapeutic applications in the treatment of neurodegenerative diseases such as Parkinson’s and Alzheimer’s [156]. Additionally, pretreated extract derived from the fungal endophytes Cytospora rhizophorae isolates HAB10R12, HAB16R13, HAB16R14, HAB16R18, and HAB8R14 obtained from Cinnamomum porrectum had inhibitory effects on the production of NO, interleukin (IL)-6, and TNF-α by activated BV2 microglia cells [157]. Furthermore, various endophytic fungi such as Aspergillus niger, Rhizopus oryzae, Dendryphion nanum, Pleospora tarda, and Penicillium sp. also showed anti-inflammatory properties. These fungi have demonstrated the ability to inhibit the activity of COX 1, COX 2, and 5-lipoxygenase which are involved in the inflammatory process. Additionally, they also produce herbarin, known for its anti-inflammatory activity. Studies also suggested that the anti-inflammatory activity of these fungi are dose-dependent, and they have been found to inhibit protein and albumin denaturation [158,159,160] (Table 4, Figure 5).

3.4. Antidiabetic Compounds from Endophytic Fungi

Diabetes mellitus (DM) is a chronic metabolic disorder marked by elevated levels of glucose in the blood (hyperglycemia) and the disruption of carbohydrate, protein, and fat metabolism. DM is linked to various complications such as cardiovascular disorders, retinopathy, nephropathy, and neuropathy. The prevalence of DM is rising, and projections indicate that by 2030 about 522 million individuals will be affected worldwide. India in particular is expected to experience a high burden of DM cases in the future. One management strategy for DM is the inhibition of α-glucosidase and α-amylase enzymes. These enzymes play a crucial role during the breakdown of carbohydrates during digestion. By slowing down their activity, the rate of carbohydrate digestion and subsequent absorption of glucose into the blood stream can be reduced, leading to better control of blood glucose levels. Inhibiting these enzymes has proven to be an effective approach in managing DM and mitigating hyperglycemia. Acarbose and miglitol are examples of drugs that specifically inhibit α-glucosidase activity, thus helping to regulate blood levels in individuals with DM. Recent studies have indicated that fungal endophytes have the potential to serve as valuable source of inhibitors for α-glucosidase and α-amylase. The compounds S (+)-2 cis 4-trans abscisic acid and 7′ hydroxyl abscisic acid, 4′ deshydroxyl, and altersolanol A isolated from endophytic Nigrospora oryzae associated with Combretum dolichopetalum demonstrated a significant reduction in blood sugar levels in mice with induced diabetes. S (+)-2 cis 4-trans abscisic acid specifically showed antidiabetic properties by enhancing the activity of peroxisome proliferator-activated gamma receptor (PPAR γ) in immune cells [173]. Thielavins A, J, and K obtained from endophytic fungal isolate MEXU 27095 exhibited a dose-dependent inhibition of α-glucosidase, with IC50 values of 15.8, 22.1, and 23.8 µM, respectively [174]. Likewise, Aspergiamides A and F, isolated from Aspergillus sp. derived from Sonneratia apetala, demonstrated α-glucosidase inhibitory activity with IC50 values of 40 and 83 µM, respectively [175]. Peptides produced by Aspergillus awamori significantly inhibited the activity of both α-glucosidase and α-amylase with IC50 values of 3.75 and 5.62 µg/mL, respectively. These inhibitors were stable over a wide range of pH and temperature conditions and exhibited nonmutagenic properties [176]. Fungal endophytes derived from medicinal diabetic plants in Uzbekistan exhibited a remarkable 60–82% inhibitory activity against α-amylase. Recently, K-10, a polymethoxylated flavone methanolic extract from endophytic Aspergillus egypticus-HT166S isolated from Helianthus tuberosus showed an inhibition of α-amylase similar to a reference standard (acarbose) in lab conditions [177,178]. Similarly, endophytic isolates from Stemphylium globuliferum PTFL005 and PTFL011 exhibited inhibitory activity against α-glucosidase with IC50 values of 17.37 and 10.71 µg/mL, respectively. Additionally, Stemphylium globuliferum PTFL005 and PTFL006 demonstrated encouraging α-amylase inhibitory activity with IC50 values of 15.48 and 13.48 µg/mL, respectively [179]. Endophytic Alternaria destruens isolated from Calotropis gigantea exhibited a weak inhibition of α-amylase (31%) and a strong inhibition of α-glucosidase (93%). Similarly, endophytic Xylariaceae sp. QGS01, Penicillium citrinum, and Colletotrichum sp. were also reported as potential inhibitors of α-glucosidase, suggesting their possible use in the management of DM [41,42,180].
Antidiabetic properties have been observed in several marine- and mangrove-derived fungi. Studies have identified certain compounds such as eremophilane sesquiterpenes from endophytic Xylaria sp., and isopimarane diterpene and 11-deoxydiapothein A from Epicoccum sp. HS-1 significantly inhibited α-glucosidase enzyme [181,182]. Similarly, tripalmitin, a mixed inhibitor derived from mangrove endophytic Zasmidium sp. strain EM5-10 exhibited significant inhibitory activity against α-glucosidase compared to acarbose. In silico studies of tripalmitin predicted that it bound to the same site as acarbose as well as an additional allosteric site in human intestinal α-glucosidase [183]. The aforementioned studies indicate that endophytes hold promise as novel inhibitors of α-amylase and α-glucosidase, which can contribute to the improved management of DM. By harnessing these endophytes, it may be possible to develop effective strategies for better control and treatment of DM (Table 5, Figure 6).

3.5. Immunosuppressive Compounds from Endophytic Fungi

Immunosuppressive medications are essential in preventing, suppressing, or minimizing organ rejection in transplant patients. As a result, they are of utmost importance in effectively managing autoimmune diseases such as lupus, psoriasis, insulin-dependent diabetes, and rheumatoid arthritis. Despite their effectiveness, these medications are associated with potential side effects, emphasizing the necessity to seek safer alternatives that can offer effective immune modulation while minimizing adverse effects [185,186]. Fungal endophytes present a promising and innovative alternative source of immunosuppressive agents and have the potential to be developed into new therapeutic drugs [187]. Recent studies have found that certain compounds of endophytic origin, such as colutellin A, dibenzofurane, lipopeptide, sydoxanthone A and B, subglutinol A and B, and 13-O-acetylsydowinin B have potent immunosuppressive properties. These findings open new possibilities for the development of novel immunosuppressive drugs. However, it is important to note that these drugs are in the early stages of investigation, and further studies are warranted to assess their safety, effectiveness, and potential side effects [5]. Two endophytic fungi (PGS1 and NLL3) isolated from Psidium guajava and Newbouldia laevis, respectively, produced citrinin, nidulalin, p-hydroxybenzoic acid, and cyclopenin. These compounds have been associated with immunosuppressant properties [188]. Similarly, a chemical analysis of endophytic fungus Mycosphaerella nawae ZJLQ129 derived from Smilax china leaves demonstrated the presence of a novel amide derivative (−)mycousnine enamine. This derivative was found to selectively inhibit T-cell proliferation by blocking the expression of surface activation antigens CD25 and CD69. These findings indicate that endophytic fungi have the potential to serve as a valuable source of immunosuppressants that exhibit a high efficacy and low toxicity [189]. Similarly, the endophytic fungus Penicillium sp. ZJ-SY2, which was found in association with the mangrove species Sonneratia apetala, produces a collection of nine polyketides that include two novel benzophenone derivatives named peniphenone and methyl peniphenone, as well as seven xanthones. These compounds demonstrated potent immunosuppressive properties, with IC50 values ranging from 5.9 to 9.3 µg/mL [190]. Endophytic Fusarium subglutinans, isolated from Tripterygium wilfordii, yielded subglutinol A and B, which have been reported to possess immunosuppressive properties [191]. Likewise, endophytic fungus Albifmbria viridis isolated from Chinese medicinal plant produced Albifpyrrols B, specifically inhibited the proliferation of B-lymphocyte cells induced by lipopolysaccharides (LPS) with an IC50 value of 16.16 µM [47]. The endophytic Phomopsis sp. S12 derived compound libertellenone J has also been found to have notable immunosuppressive properties. It effectively reduces the production of NO, IL-1β, IL-6, and TNF-α with IC50 values ranging from 2.2 to 10.2 µM. In addition, it also decreases the expression of iNOS, IL-1β, IL-6, and TNF-α mRNA in LPS-activated macrophages, with IC50 values ranging from 3.2 to 15.2 µM [192]. Furthermore, a fermentation extract of endophytic Botryosphaeria dothidea BAK-1 isolated from Kigelia africana demonstrated a dose-dependent suppression of T-cell proliferation by 50% and TNF-α production by 55% [193]. These significant reports inspire the further exploration of fungal endophytes for new immunosuppressive agents [192] (Table 6, Figure 7).

3.6. Antimicrobial Compounds from Endophytic Fungi

The emergence of drug resistance among disease causing microorganisms is a burgeoning issue that needs urgent action. Infectious diseases are among the leading causes of deaths after cardiovascular disorders and cancers, as they account for 13.7 million deaths globally (13.6% of total global deaths) (Institute of Health Metrics and Evaluation 2019). The COVID-19 outbreak is a prime example of this situation, caused by the spread of a novel coronavirus. This virus has infected over 600 million individuals and tragically caused the death of more than 6.5 million people across the globe [1]. To address this pressing issue, there is a continuous quest to discover novel antimicrobial agents that are both effective and have reduced or minimal side effects. Endophytic fungi have been well recognized for their ability to produce a diverse array of secondary metabolites such as alkaloids, terpenoids, flavonoids, and polyketides. These compounds have demonstrated antimicrobial activity against various pathogenic microorganisms such as Staphylococcus aureus, Pseudomonas aeruginosa, Klebsiella pneumonia, Shigella flexneri, Enterococcus faecalis, Escherichia coli, Salmonella typhi, Bacillus subtilis, Saccharomyces cerevisiae, Candida albicans, F. oxysporum, human immunodeficiency virus (HIV), herpes simplex virus (HSV) and influenza virus (H1N1) [198,199,200,201,202,203]. In recent years, numerous bioactive metabolites have been isolated from endophytic fungi, exhibiting profound antimicrobial activities. Table 7 provides a comprehensive overview of these antimicrobial agents, highlighting their antibacterial, antifungal, and antiviral properties. Fumigaclavine C and fraxetin produced by A. fumigatus obtained from Ceriops decandra exhibited strong antibacterial activity against E. coli, Micrococcus luteus, S. aureus, and P. aeruginosa [204]. Antibacterial activity has been observed in Cristatumin B, quiannulatic acid, and Dihydroauroglaucin, which were isolated from endophytic Aspergillus niger and Emericella sp. These compounds have exhibited broad-spectrum activity against pathogenic bacteria such as E. faecalis, K. pneumonia, P. aeruginosa, and multidrug resistant Staphylococcus aureus (MDRSA) [205,206]. Similarly, endophytic Athelia rolfsii, isolated from Coleus amboinicus produced an aromatic compound containing methoxy, hydroxyl, and methyl groups that exhibited strong antibacterial activity against B. subtilis, E. coli, P. aeruginosa, S. aureus, and Streptococcus mutans [207]. Additionally, Penicillium citrinum isolated from Digitaria bicornis secreted ciprofloxacin, which displayed antibacterial activity against E. coli, E. faecalis, S. aureus, and S. typhi [203] (Table 7, Figure 8).
7-Hydroxycoumarine, β-asarone, diphenyl sulfone, and griseofulvin, produced by Curvularia protuberate isolated from Paspalidium favidum demonstrated antifungal activity against Alternaria alternata and F. oxysporum with an IC50 values of 31 and 62 µg/mL, respectively [203]. 3-phenylpropionic acid derived from endophytic Cladosporium cladosporioides isolated from Zygophyllum mandavillei displayed antifungal properties towards Aspergillus flavus and F. solani with IC50 values of 3.9 and 15.62 mg/mL [234]. Aplojaveediins A-F extracted from endophytic fungus Aplosporella javeedii found in association with Orychophragmus violaceus exhibited notable activity against C. albicans ATCC24433 [235]. Similarly, endophytic F. oxysporum KU527806 isolated from Dendrobim lindley synthesized Gibepyrone A, Pyrrolo[1,2-a]pyrazine-1, 4-dione, hexahydro-3-(2-methylpropyl) and indole acetic acid which demonstrated significant inhibitory activity against C. albicans, Candida tropicalis, Curvularia, and Fusarium species [237]. Similarly, endophytic Lophodermium nitens DAOM 250027 isolated from Pinus strobus produces (7R)-(-)-methoxysydonol and its derivatives (7R,7′R)-(-)-pyrenophorin, which showed antifungal activity against S. cerevisiae [199]. Furthermore, Phialomustin C and D isolated from endophytic Phialophora mustea in Crocus sativus exhibited antifungal activity against C. albicans with an IC50 values of 14.3 and 73.6 µM, respectively [241] (Table 7, Figure 9).
Endophytic Acremonium sp. MER V1 and Chaetomium sp. MER V7 isolated from Avicennia marina showed antiviral activity against hepatitis C virus. However, their fusant MER V6270 showed a stronger inhibition of hepatitis C virus as compared to individual fungus Acremonium sp. MER V1 and Chaetomium sp. MER V7 [257]. Phomanolide B obtained from endophytic Phoma sp. demonstrated antiviral properties towards influenza virus, whereas a novel bioactive compound Aspulvinone E, obtained from endophytic fungi A. terreus displayed strong antiviral activity against HIV [245,246]. Pestalotiopsis thea is an endophytic fungus that produces bioactive metabolites such as chloroisosulochrin, ficipyrone A and pestheic acid. Amongst them, chloroisosulochrin displayed maximum antirespiratory syncytial viral (RSV) inhibitor activity, whereas the other two compounds exhibited moderate activities against the virus [250]. Endophytic Pleospora tarda secreted alternariol and alternariol–(9)-methyl ester that showed moderate inhibitory activity against HSV (40%). Furthermore, fungal endophytes such as Nigrospora sphaerica, Acremonium strictum, Phoma leveillei, Aspergillus flavus, Chaetomium globosum, Mucor fuscus, Acremonium strictum, and Penicillium chrysogenoum, which were isolated from Chiliadenus montanus, Launea spinosa, Euphorbia sancta, Stachys aegyptiaca, Hypericum sinaicum, Stachys aegyptiaca, and Launea spinose, respectively, displayed weak to moderate (2–14%) activity against HSV [145]. COVID-19 is a new viral pandemic disease that originated in China and has spread to all countries worldwide. Currently, there is no specific drug available to treat COVID-19, and management is mainly focused on supportive care such as vitamin supplements, antibiotics, and oxygen therapy. Some researchers have proposed the possibility that endophytes may possess antiviral properties that could be effective against novel coronaviruses. In a study by [208], it was found that crude ethyl acetate extract derived from endophytic Curvularia papendorfii demonstrated potent antiviral activity against human coronavirus HCoV229E and feline coronavirus FCV F9. Furthermore, in another study, it was observed that fungal endophytes produced Aspergillide B1 and 3a-Hydroxy-3,5-dihydromonacolin L compounds. These compounds exhibited the highest binding energy scores when interacting with the protease (Mpro) of the novel coronavirus, indicating their potential as inhibitors against the virus [245]. However, it is crucial to emphasize that these findings are derived from preclinical studies and additional research is necessary to validate their effectiveness in in vivo settings and establish optimal dosage and administration protocols. Additionally, conducting clinical trials would be necessary to assess the safety and efficacy of fungal endophytes as a potential bioresource in the treatment of COVID-19 (Table 7, Figure 10).

3.7. Antiprotozoal Compounds from Endophytic Fungi

Protozoan parasites such as Tryanosoma cruzi, Plasmodium berghei, Plasmodium falciparum, and Leishmania amazonensis cause a range of diseases including Chagas disease, malaria and leishmaniasis. These diseases are vector-borne and are transmitted to humans through the bite of infected mosquitoes or flies [258]. They are classified as neglected diseases by the WHO, and primarily affect low-income areas, receiving limited attention in terms of research and development. In addition, the current drugs available for treating these diseases have significant limitations such as poor effectiveness, toxicity, drug resistance, and high cost. As a result, there is an urgent need to find new drugs that are effective, safer, and affordable. To address these issues, efforts are being made to explore different strategies such as repurposing existing drugs, screening chemical libraries, and developing new candidates through targeted or natural product-based approaches [259]. Studies suggest that the endophytic fungi derived from medicinal plants such as Artemisia annua, Cinchona calisaya, and Markhamia platycalyx have been found to produce bioactive compounds with inhibitory properties against the above-mentioned parasites. Notably, endophytic Nigrospora oryzae Cf-298113, isolated from the roots of Triticum sp., secrete pipecolisporin, which has potent inhibitory activity against P. falciparum (3.21 µM) and T. cruzi (8.68 µM) [260]. The antiplasmodial activity of endophytic Aspergillus terrus, Penicillum commune, P. chrysogenum, and Talaromyces piophilus isolated from A. annua has been investigated. Among these, the fermentation extract of P. commune and P. chrysogenum inhibited P. falciparum with IC50 values of 1.1 and 3.3 µg/mL, respectively. The extract from Talaromyces strains showed a moderate activity with IC50 values of 7.6–9.9 µg/mL, whereas the extract from A. terreus displayed a lower activity with an IC50 of 35 µg/mL [261]. In addition, two endophytic fungal strains (IP-2 and IP-6) isolated from A. annua demonstrated antiplasmodial activity with IC50 values of 30 and 42 µg/mL, respectively, whereas 19,20 epoxycytochalasin C derived from the ethyl acetate extract of endophytic Nemania sp. UM10M showed a relatively weak antiplasmodial activity [262,263]. In addition, endophytic P. citrinum AMrb11 and Neocosmospora rubicola AMb22 exhibited potent antiplasmodial activity against both chloroquine-sensitivePf3D7 and chloroquine-resistant PfINDO/PfDd2 strains of P. falciparum, with IC50 values ranging from 0.39 to 1.92 µg/mL for Neocosmospora rubicola AMb22 and 0.84–0.93 µg/mL for P. citrinum Amrb11 [264]. Moreover, a fermentation extract of endophytic Aspergillus flocculus yielded 3-hydroxymellein and dorcinol, which demonstrated significant inhibitory effects of 56 and 97% against the sleeping-sickness-causing parasite T. cruzi. The antitrypanosomal activity of A. flocculus is believed to be attributed to the synergistic effects of active steroidal compounds such as campesterol, ergosterol, and ergosterol peroxide [265]. Similarly, lead extracts obtained from endophytic isolates sourced from Antarctic angiosperms, particularly Deschampsia antartica, were tested for their ability to inhibit the proliferation of L. amazonensis. The IC50 values of these extracts ranged from 0.2 to 125 µg/mL. Notably, Alternaria, Cadophora, Herpotrichia, and Phaeosphaeria spp. exhibited over 90% killing of L. amazonensis [266]. Recently, an in silico approach was employed to investigate the antileishmanial activity of epicoccamide derivatives A–D, which are of endophytic origin. These derivatives interacted with the active site of the enzyme through hydrogen bonds and hydrophobic interactions, leading to their stabilization. Epicoccamide derivatives exhibited high bonding energies with the trypanothione reductase of −13.31, −13.44, −13.31, and −13.32 kcal/mol, respectively [267,268] (Table 8, Figure 11).

4. Prospects and Challenges

Over the last few years, endophytic fungi have attracted significant attention in natural-product-based drug discovery due to their inherent capability to produce secondary metabolites as a source of novel drugs with low toxicity for treating various human ailments [33,34]. However, despite the progress made in studying endophytic fungi, only a fraction of endophytes have been explored so far (about 1%), and the vast majority of these organisms remain untapped and uncharacterized, with great potential for discovering new bioactive compounds [5,9,23,37,52]. To effectively isolate endophytes with significant bioactivity, a selection of host plant and its ecological niche is crucial. Plants that inhabit areas with high biodiversity, particularly those with endemic plant species, are more likely to harbor endophytes with novel chemical entities. When selecting a host plant for endophyte isolation, preference should be given to plants with known medicinal properties. This approach enhances the likelihood of identifying endophytes that produce bioactive compounds relevant to human health [4,34,48,51]. Furthermore, establishing connections between fungal metabolites and plant genomics enhances our understanding of the biosynthetic pathways involved in the process, justifying the production of the metabolites based on scientific knowledge and evidence, rather than relying upon unproven hypotheses [1,5,18,22]
The production of bioactive compounds from fungal endophytes on an industrial scale is a complex and arduous task, necessitating advanced and efficient approaches [23]. Cutting-edge techniques such as CRISPR-Cas9 and epigenetic modifiers show promise in enhancing bioactive compound production. Moreover, several other strategies such as optimizing culture parameters, employing elicitors, and utilizing coculture fermentation have been successfully employed in laboratory conditions to augment the production of bioactive compounds from fungal endophytes [43]. However, isolating and characterizing promising fungal endophytes capable of producing bioactive compounds has always posed significant challenges. The integration of molecular approaches and bioinformatics, including phylogenetic studies, offers a potential solution by facilitating the precise delineation of fungal strains at the species level [23]. Under in situ conditions, endophytes coexist and interact with various other organisms, which significantly influences the production of secondary metabolites. However, when studied in in vitro conditions, endophytes are typically cultured under axenic conditions, devoid of these natural interactions. Therefore, it is essential to explore the interactions among endophytes, their host plants, and other associated microorganisms to fully harness their potential for the production of bioactive compounds [21,32,37]. These interactions are highly sensitive to culture conditions, offering an opportunity to optimize in vitro conditions and create an environment that stimulates the production of the desired bioactive compounds [4,9,34]. By adjusting culture conditions, media composition, aeration rate, and temperature, it is feasible to produce a specific desired compound. Furthermore, cocultivating endophytes in the presence of other microorganisms triggers the activation of biosynthetic pathways, leading to the synthesis of bioactive metabolites which are not produced when endophytes are cultured individually. Consequently, extensive research will be necessary to gain a comprehensive understanding of endophytes’ biosynthetic capabilities. By developing suitable cocultivation methods and optimizing culture conditions, a consistent and efficient production of desired bioactive compounds may be possible from endophytic fungi in the future [18,21,43].
The process of discovering natural products traditionally involves bioprospecting various organisms and conducting laboratory screening programs, resulting in complex data. However, this approach often faces high attrition rates and challenges. To overcome these issues, artificial intelligence (AI) and machine language (ML) are increasingly being employed. The recent breakthroughs in AI, particularly in ML, have revolutionized the field of natural-product-based drug discovery programs. AI tools have demonstrated their effectiveness in uncovering hidden patterns, classifying objects, and clustering compounds based on their characteristics [280,281]. AI tools such as LeafNet, LeafSnap, ResNet26, IDBac, and SPeDE have been developed to assist in taxonomic identification, enabling the selection of novel organisms. For genome mining and chemical dereplication [282,283,284,285,286], AI tools such as ANtiSMASH, MIBiG, IMG-ABC, NRPro, CHEM, ELINA, and DEREP-NP have proven valuable. These tools help in the analysis and interpretation of genomic data, allowing researchers to identify potential gene clusters responsible for the biosynthesis of bioactive compounds. Furthermore, they aid the dereplication process by comparing chemical structures and identifying known compounds, thereby facilitating the selection of novel organisms with unique chemical profiles [287,288,289,290,291,292,293]. In the field of target identification, AI tools such as AutoDock, Schrodinger, SDiDER, and BANDIT play a crucial role. These tools utilize molecular docking and ligand-based approaches to predict the interactions between bioactive compounds and target proteins. By simulating the binding process, potential targets can be identified and the design of new compounds optimized [294,295,296,297]. The integration of AI tools into bioactive compound discovery has significantly enhanced the efficiency and accuracy of the process, accelerating the identification and development of bioactive compounds with therapeutic potential [280].

5. Conclusions

The microbial world of plants holds great promise for future medicine. The scientific community has directed considerable attention towards fungal endophytes, recognizing their potential to synthesize bioactive compounds with a wide range of properties that may be antimicrobial, anticancer, antioxidant, anti-inflammatory, antidiabetic, immunomodulatory, and cardio-protective. This underscores that fungal endophytes are a bioresource for the development of novel drugs and other biotechnology products. Studies have shown that a significant portion (about 51%) of the bioactive metabolites sourced from endophytic fungi possess unique chemical structures. This emphasizes the existence of a vast and untapped reservoir, holding great potential for future exploration and development. The field of fungal endophytic biology has experienced significant technological advancement that has opened fresh avenues for the isolation and characterization of novel bioactive compounds. These advances encompass sophisticated molecular techniques to isolate and characterize endophytic fungi, as well as the development of novel methods to isolate bioactive compounds, both culture-dependent and culture-independent. These modern methods have greatly enhanced the efficacy and precision of isolation processes, enabling the discovery of previously unknown bioactive compounds from endophytic fungi. Moreover, the integration of bioinformatics tools and computational biology approaches has played a pivotal role in the discovery and characterization of bioactive compounds from endophytic fungi. These tools have provided valuable insights into the biosynthesis and regulation of secondary metabolites within endophytic fungi, facilitating the identification of new gene clusters and biosynthetic pathways associated with bioactive compound production. By leveraging these technological advances, researchers are now able to delve deeper into the untapped potential of endophytic fungi and uncover a wealth of promising bioactive compounds. However, despite the advancements in the field, the exploration of endophytic fungi for bioactive compounds is still in its early stages. Consequently, there is a pressing need to realign our research priorities towards biotechnological advances to expedite the screening and discovery of new biomolecules. However, conducting a thorough review of the literature and documentation regarding host plants, biosynthetic machineries, and their mechanism of action can yield valuable insights for potential explorations and bioprospecting endeavors. This comprehensive understanding offers opportunities to harness endophytic fungi as a sustainable and renewable source of bioactive compounds, contributing to human health and addressing the challenges of antibiotic resistance.

Author Contributions

A.G. and V.M. wrote the main manuscript; M.G., S.G. and K.A.Q. prepared figures; K.K.S. and M.J. conceived the idea and arranged funds. All authors have read and agreed to the published version of the manuscript.

Funding

The research was funded by Department of Biotechnology (DBT) under project no. BT/PR29526/FcB/125/16/2018. MJ would like to acknowledge the King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia for financial support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

A.G. is thankful to Pt. Ravishankar Shukla University, Raipur, C.G. India for providing Pt. Ravishankar Shukla University Research Scholarship 2020-21. K.S. is thankful to the Department of Biotechnology (DBT) for sponsoring the project no. BT/PR29526/FcB/125/16/2018. MJ would like to acknowledge the King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia for financial support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Caruso, D.J.; Palombo, E.A.; Moulton, S.E.; Zaferanloo, B. Exploring the promise of endophytic fungi: A review of novel antimicrobial compounds. Microorganisms 2022, 10, 1990. [Google Scholar] [CrossRef] [PubMed]
  2. Pasrija, P.; Girdhar, M.; Kumar, M.; Arora, S.; Katyal, A. Endophytes: An unexplored treasure to combat Multidrug resistance. Phytomed. Plus 2022, 2, 100249. [Google Scholar] [CrossRef]
  3. Digra, S.; Nonzom, S. An insight into endophytic antimicrobial compounds: An updated analysis. Plant Biotechnol. Rep. 2023, 1–31. [Google Scholar] [CrossRef]
  4. Deshmukh, S.K.; Verekar, S.A.; Bhave, S.V. Endophytic fungi: A reservoir of antibacterials. Front. Microbiol. 2015, 5, 715. [Google Scholar] [CrossRef] [Green Version]
  5. Adeleke, B.S.; Babalola, O.O. Pharmacological potential of fungal endophytes associated with medicinal plant: Review. J. Fungi 2021, 7, 147. [Google Scholar] [CrossRef] [PubMed]
  6. Cragg, M.G.; Newman, D.J. Natural products: A continuing source of novel drug leads. BBA Gen. Subj. 2013, 1830, 3670–3695. [Google Scholar] [CrossRef] [Green Version]
  7. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs from 1981 to 2014. J. Nat. Prod. 2016, 79, 629–661. [Google Scholar] [CrossRef] [Green Version]
  8. Cruz, J.S.; Silva, C.A.D.; Hamerski, L. Natural products from endophytic fungi associated with Rubiaceae species. J. Fungi 2020, 6, 128. [Google Scholar] [CrossRef]
  9. Kaul, S.; Gupta, S.; Ahmed, M.; Dhar, M.K. Endophytic fungi from medicinal plants: A treasure hunt for bioactive metabolites. Phytochem. Rev. 2012, 11, 487–505. [Google Scholar] [CrossRef]
  10. Abdel-Azeem, A.M.; Azeem, M.A.A.; Hadi, S.Y.A.; Darwish, A.G. Aspergillus: Biodiversity, ecological significances, and industrial applications. In Recent Advancement in White Biotechnology through Fungi Diversity and Enzymes Perspectives; Yadav, A., Mishra, S., Singh, S., Gupta, A., Eds.; Springer: Cham, Switzerland, 2019; Volume 1, pp. 121–179. [Google Scholar]
  11. Chandra, S. Endophytic fungi: Novel sources of anticancer lead molecules. Appl. Microbiol. Biot. 2012, 95, 47–59. [Google Scholar] [CrossRef]
  12. Kumar, S.; Aharwal, R.P.; Jain, R.; Sandhu, S.S. Bioactive molecules of endophytic fungi and their potential in anticancer drug development. Curr. Pharmacol. Rep. 2021, 7, 27–41. [Google Scholar] [CrossRef]
  13. Tavares, D.G.; Barbosa, B.V.L.; Ferreira, R.L.; Duarte, W.F.; Cardoso, P.G. Antioxidant activity and phenolic compounds of the extract from pigment producing fungi isolated from Brazilian caves. BIocatat. Agric. Biotechnol. 2018, 16, 148–154. [Google Scholar] [CrossRef]
  14. Dudka, M.M.; Jaszek, M.; Blachowicz, A.; Rejczak, T.P.; Matuszewska, A.; Jaroszuk, M.O.; Stefaniuk, D.; Janusz, G.; Sulej, J.; Szerszen, M.K. Fungus Cerrena unicolor as an effective source of new antiviral, immunomodulatory, and anticancer compounds. Int. J. Biol. Macromol. 2015, 79, 459–468. [Google Scholar] [CrossRef]
  15. Hawksworth, D.L.; Rossman, A.Y. Where are all the undescribed fungi? Phytopathology 1997, 87, 888–891. [Google Scholar] [CrossRef] [Green Version]
  16. UI-Hassan, U.R.; Strobel, G.A.; Booth, E.; Knighton, B.; Floerchinger, C.; Sears, J. Modulation of volatile organic compound formation in the mycodiesel-producing endophyte Hypoxylon sp. CI-4. Microbiology 2012, 158, 465–473. [Google Scholar] [CrossRef] [Green Version]
  17. Qadri, M.; Johri, S.; Shah, B.A.; Khajuriya, A.; Sidiq, T.; Lattoo, S.K.; Abdin, M.Z.; Riyaz-UI-Hassan, S. Identification and bioactive potential of endophytic fungi isolated from selected medicinal plants of the Western Himalayas. SpringerPlus 2013, 2, 8. [Google Scholar] [CrossRef] [Green Version]
  18. Deshmukh, S.K.; Gupta, M.K.; Prakash, V.; Saxena, S. Endophytic fungi: A source of potential antifungal compounds. J. Fungi 2018, 4, 77. [Google Scholar] [CrossRef] [Green Version]
  19. Chowdhary, K.; Kaushik, N.; Coloma, A.G.; Raimundo, C.M. Endophytic fungi and their metabolites isolated from Indian medicinal plant. Phytochem. Rev. 2012, 11, 467–485. [Google Scholar] [CrossRef]
  20. Meshram, V.; Kapoor, N.; Dwibedi, V.; Srivastava, A.; Saxena, S. Extracellular resveratrol producing endophytic fungus, Quambalaria cyanescens. S. Afr. J. Bot. 2022, 146, 409–416. [Google Scholar] [CrossRef]
  21. Khare, E.; Mishra, J.; Arora, N.V. Multifaceted interactions between endophytes and plant: Developments and prospects. Front. Microbiol. 2018, 9, 2732. [Google Scholar] [CrossRef]
  22. Strobel, G. The emergence of endophytic microbes and their biological promise. J. Fungi 2018, 4, 57. [Google Scholar] [CrossRef] [Green Version]
  23. Meshram, V.; Gupta, M. Endophytic fungi: A quintessential source of potential bioactive compounds. In Sustain: Endophytes for a Growing World; Hodkinson, T., Doohan, F., Saunders, M., Murphy, B., Eds.; Cambridge University Press: Cambridge, UK, 2019; Volume 13, pp. 277–309. [Google Scholar]
  24. Meshram, V.; Elazar, M.; Maymon, M.; Sharma, G.; Shawahna, R.; Charuvi, D.; Freeman, S. Endophytic Fusarium clavum confers growth and salt tolerance in Cucumis melo. Environ. Exp. Bot. 2023, 206, 105–153. [Google Scholar] [CrossRef]
  25. Aly, A.H.; Debbab, A.; Proksch, P. Fungal endophytes—Secret producers of bioactive plant metabolites. Int. J. Pharm. Sci. 2013, 68, 499–505. [Google Scholar]
  26. Saxena, S.; Meshram, V.; Kapoor, N. Musocdor tigerii sp. non-volatile antibiotic producing endophytic fungus from the Northeatern Himalayas. Ann. Microbiol. 2015, 65, 47–57. [Google Scholar] [CrossRef]
  27. Venugopalan, A.; Srivastava, S. Endophytes as in vitro production platforms of high value plant secondary metabolites. Biotechnol. Adv. 2015, 33, 873–887. [Google Scholar] [CrossRef] [PubMed]
  28. Kusari, S.; Spiteller, M. The promise of endophytic fungi as sustainable resource of biologically relevant pro-drugs: A focus on Cameroon. In Fungi Applications and Management Strategies; CRC Press: Boca Raton, FL, USA, 2016; pp. 1–13. [Google Scholar]
  29. Krohn, K.; Ullah, Z.; Hussain, H.; Florke, U.; Schulz, B.; Draeger, S.; Pescitelli, G.; Salvadori, P.; Antus, S.; Kurtan, T. Massarilactones e-g, new metabolites from the endophytic fungus Coniothyrium sp., associated with the plant Artimisia maritime. Chirality 2007, 19, 464–470. [Google Scholar] [CrossRef] [PubMed]
  30. Kusari, S.; Hertweck, C.; Spiteller, M. Chemical ecology of endophytic fungi: Origins of secondary metabolites. Chem. Biol. 2012, 19, 792–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Howitz, K.T.; Sinclair, D.A. Xenohormesis: Sensing the chemical cues of other spices. Cell 2008, 133, 387–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kusari, S.; Singh, S.; Jayabaskaran, C. Biotechnological potential of plant-associated endophytic fungi: Hope versus hype. Trends Biotechnol. 2014, 32, 297–303. [Google Scholar] [CrossRef] [PubMed]
  33. Bedi, A.; Adholeya, A.; Deshmukh, S.K. Novel anticancer compounds from endophytic fungi. Curr. Biotechnol. 2018, 7, 168–184. [Google Scholar] [CrossRef]
  34. Deshmukh, S.K.; Gupta, M.K.; Prakash, V.; Reddy, M.S. Fungal endophytes: A novel source of cytotoxic compounds. In Endophytes and Secondary Metabolites; Reference Series in Phytochemistry; Jha, S., Ed.; Springer: Cham, Switzerland, 2019; pp. 365–426. [Google Scholar]
  35. Li, H.L.; Li, X.M.; Li, X.; Wang, C.Y.; Liu, H.; Kassack, M.U.; Meng, L.H.; Wang, B.G. Antioxidant hydroanthraquinones from the marine algal-derived endophytic fungus Talaromyces islandicus EN-501. J. Nat. Prod. 2016, 80, 162–168. [Google Scholar] [CrossRef]
  36. Kaur, N.; Arora, D.S.; Kalia, N.; Kaur, M. Antibioflm, antiproliferative, antioxidant and antimutagenic activities of an endophytic fungus Aspergillus fumigatus from Moringa oleifera. Mol. Biol. Rep. 2020, 47, 2901–2911. [Google Scholar] [CrossRef]
  37. Manganyi, M.C.; Ateba, C.N. Untapped potentials of endophytic fungi: A review of novel bioactive compounds with biological applications. Microorganisms 2020, 8, 1934. [Google Scholar] [CrossRef]
  38. Nischitha, R.; Shivanna, M.B. Screening of secondary metabolites and antioxidant potential of endophytic fungus Penicillium citrinum and host Digitaria bicornis by spectrophotometric and electrochemical methods. Arch. Biol. 2022, 204, 206. [Google Scholar] [CrossRef]
  39. Pal, P.P.; Shaik, A.B.; Begum, A.S. Prospective leads from endophytic fungi from anti-inflmmatory drug discovery. Planta Med. 2020, 86, 941–959. [Google Scholar] [PubMed] [Green Version]
  40. Mazumder, K.; Ruma, Y.N.; Akter, R.; Aktar, A.; Hossain, M.M.; Shahina, Z.; Mazumdar, S.; Kerr, P.G. Identification of bioactive metabolites and evaluation of in vitro anti-inflammatory and in vivo antinociceptive and antiarthritic activities of endophyte fungi isolated from Elaeocarpus floribundus blume. J. Ethnopharmacol. 2021, 273, 113975. [Google Scholar] [CrossRef]
  41. Artanti, N.; Tachibana, S.; Kardono, L.B.; Sukiman, H. Isolation of alpha-glucosidase inhibitors produced by an endophytic fungus, Colletotrichum sp. TSC13 from Taxus sumatrana. Pak. J. Biol. Sci. 2012, 15, 673–679. [Google Scholar] [CrossRef] [Green Version]
  42. Indrianingsih, A.W.; Tachibana, S. α-Glucosidase inhibitor produced by an endophytic fungus, Xylariaceae sp. QGS 01 from Quercus gilva Blume. Food Sci. Hum. Wellness 2017, 6, 88–95. [Google Scholar] [CrossRef]
  43. Agrawal, S.; Samanta, S.; Deshmukh, S.K. The antidiabetic potential of endophytic fungi: Future prospects as therapeutic agents. Biotechnol. Appl. Biochem. 2021, 69, 1159–1165. [Google Scholar] [CrossRef] [PubMed]
  44. Song, X.Q.; Zhang, X.; Han, Q.J.; Li, X.B.; Li, G.; Li, R.J.; Jiao, Y.; Zhou, J.C.; Lou, H.X. Xanthone derivatives from Aspergillus sydowii, an endophytic fungus from the liverwort Scapania ciliata S. Lac and their immunosuppressive activities. Phytochem. Lett. 2013, 6, 318–321. [Google Scholar] [CrossRef]
  45. Zhang, W.; Li, J.; Wei, C.; Deng, X.; Xu, J. Chemical epigenetic modifiers enhance the production of immunosuppressants from the endophytic fungus Aspergillus fumigatus isolated from Cynodon dactylon. Nat. Prod. Res. 2021, 36, 4481–4485. [Google Scholar] [CrossRef] [PubMed]
  46. Deng, M.; Chen, X.; Shi, Z.; Xie, S.; Qiao, Y.; Chen, G.; Tan, X.; Lu, Y.; Qi, C.; Zhang, Y. New immunosuppressive secondary metabolites from the endophytic fungus Aspergillus sp. Fitoterapia 2021, 151, 104882. [Google Scholar] [CrossRef] [PubMed]
  47. Wei, P.P.; Ji, J.C.; Ma, X.J.; Li, Z.H.; Ai, H.L.; Lei, X.X.; Liu, J.K. Three new pyrrole alkaloids from the endophytic fungus Albifmbria viridis. Nat. Prod. Bioprospect. 2022, 12, 5. [Google Scholar] [CrossRef]
  48. Gupta, M.; Meshram, V. The biological promises of endophytic Muscodor species. In Fungi and Their Role in Sustainable Development: Current Perspectives; Gehlot, P., Singh, J., Eds.; Springer: Cham, Switzerland, 2018; pp. 51–74. [Google Scholar]
  49. Petrini, O. Fungal endophytes of tree leaves. In Microbial Ecology of Leaves; Andrews, J.H., Hirano, S.S., Eds.; Brock/Springer Series in Contemporary Bioscience; Springer: New York, NY, USA, 1991; pp. 179–197. [Google Scholar]
  50. Bacon, C.W.; White, J.F. Physiological adaptations in the evolution of endophytism in the clavicipitaceae. In Microbial Endophytes; Bacon, C.W., White, J.F., Eds.; CRC Press: Boca Raton, FL, USA, 2000; pp. 1–26. [Google Scholar]
  51. Strobel, G.; Daisy, B. Bioprospecting for microbial endophytes and their natural products. Microbiol. Mol. Biol. Rev. 2003, 67, 491–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kusari, S.; Pandey, S.P.; Spiteller, M. Untapped mutualistic paradigms linking host plant and endophytic fungal production of similar bioactive secondary metabolites. Phytochem 2013, 91, 81–87. [Google Scholar] [CrossRef]
  53. Hodkinson, T.R. Evolution and taxonomy of the grasses (poaceae): A model family for the study of species-rich groups. Annu. Plant Rev. 2018, 15, 255–294. [Google Scholar]
  54. Schulz, B.; Boyle, C. The endophytic continuum. Mycol. Res. 2005, 109, 661–686. [Google Scholar] [CrossRef] [Green Version]
  55. De-Berry, A. Morphologie und Physiologie der Pilze, Flechten und Myxomyceten; Series: Leipzig: Hofmeister’s Handbook of Physiological Botany; Hansebooks: Norderstedt, Germany, 1866; Volume 2. [Google Scholar]
  56. Freeman, E.M. The seed-fungus of Lolium temulentum, L., the darnel. Proc. R. Soc. Lond. 1904, 71, 467–476. [Google Scholar]
  57. Stierle, A.A.; Stierle, D.B. Bioactive secondary metabolites produced by the fungal endophytes of conifers. Nat. Prod. Commun. 2015, 10, 1671–1682. [Google Scholar] [CrossRef] [Green Version]
  58. Meshram, V.; Saxena, S.; Paul, K. Xylarinase: A novel clot busting enzyme from an endophytic fungus Xylaria curt. J. Enzym. Inhib. Med. Ch. 2016, 31, 1502–1511. [Google Scholar] [CrossRef] [Green Version]
  59. Promputtha, I.; Lumyong, S.; Dhanasekaran, V.; Mc-Kenzie, E.H.C.; Hyde, K.D.; Jeewon, R. A phylogenetic evolution of whether endophytes become saprotrophs at host senescence. Microb. Ecol. 2007, 53, 579–590. [Google Scholar] [CrossRef]
  60. Tan, X.; Zhou, Y.; Zhou, X.; Xia, X.; Wei, Y.; He, L.; Tang, H.; Yu, L. Diversity and bioactive potential of culturable fungal endophytes of Dysosma versipellis; a rare medicinal plant endemic to China. Sci. Rep. 2018, 8, 5929. [Google Scholar] [CrossRef]
  61. Kusari, S.; Zuhlke, S.; Spiteller, M. An endophytic fungus from Camptotheca acuminata that produces camptothecin and analogues. J. Nat. Prod. 2009, 72, 2–7. [Google Scholar] [CrossRef]
  62. Stierle, A.; Strobel, G.; Stierle, D. Taxol and taxane production by Taxomyces andreanae, an endophytic fungus of pacific yew. Science 1993, 260, 214–216. [Google Scholar] [CrossRef] [PubMed]
  63. Stierle, A.; Strobel, G. The search for a taxol-producing microorganism among the endophytic fungi of the pacific yew, Taxus brevifolia. J. Nat. Prod. 1995, 58, 1315–1324. [Google Scholar] [CrossRef]
  64. Kharwar, R.N.; Mishra, A.; Gond, S.K.; Stierle, A.; Stierle, D. Anticancer compounds derived from fungal endophytes: Their importance and future challenges. Nat. Prod. Rep. 2011, 28, 1208–1288. [Google Scholar] [CrossRef]
  65. Nicoletti, R.; Fiorentino, A. Plant bioactive metabolites and drugs produced by endophytic fungi of spermatophyta. Agriculture 2015, 5, 918–970. [Google Scholar] [CrossRef] [Green Version]
  66. Guo, B.; Li, H.; Zhang, L. Isolation of the fungus producing vinblastine. J. Yunnan Univ. (Nat. Sci.) 1998, 20, 214–215. [Google Scholar]
  67. Puri, S.C.; Verma, V.; Amna, T.; Qazi, G.N.; Spiteller, M. An endophytic fungus from Nothapodytes foetida that produces camptothecin. J. Nat. Prod. 2005, 68, 1717–1719. [Google Scholar] [CrossRef]
  68. Eyeberger, A.L.; Dondapati, R.; Porter, J.R. Endophytes fungla isolateds from Podophyllum peltatum produce podophyllotoxin. J. Nat. Prod. 2006, 69, 1121–1124. [Google Scholar] [CrossRef]
  69. Kusari, S.; Lamshoft, M.; Zuhlke, S.; Spiteller, M. An endophytic fungus from Hypericum perforatum that produces hypericin. J. Nat. Prod. 2008, 71, 159–162. [Google Scholar] [CrossRef] [PubMed]
  70. Porras-Alfaro, A.; Bayman, P. Hidden fungi, emergent properties: Endophytes and microbiomes. Annu. Rev. Phytopathol. 2011, 49, 291–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Xu, K.; Li, X.Q.; Zhao, D.L.; Zhang, P. Antifungal secondary metabolites produced by the fungal endophytes: Chemical diversity and potential use in the development of biopesticides. Front. Microbiol. 2021, 12, 689527. [Google Scholar] [CrossRef] [PubMed]
  72. Aly, A.H.; Debbab, A.; Proksch, P. Fungal endophytes: Unique plant inhabitants with great promises. Appl. Microbiol. Biot. 2011, 90, 1829–1845. [Google Scholar] [CrossRef] [PubMed]
  73. Meshram, V.; Saxena, S.; Paul, K.; Gupta, M.; Kapoor, N. Production, purification and characterization of a potential fibrinolytic protease from endophytic Xylaria curta by solid substrate fermentation. Appl. Biochem. Biotech. 2016, 181, 1496–1512. [Google Scholar] [CrossRef] [PubMed]
  74. Zilla, M.K.; Qadri, M.; Pathania, A.S.; Strobel, G.A.; Nalli, Y.; Kumar, S.; Guru, S.K.; Bhushan, S.; Singh, S.K.; Vishwakarma, R.A.; et al. Bioactive metabolites from an endophytic Cryptosporiopsis sp. inhabiting Clidemia hirta. Phytochemistry 2013, 95, 291–297. [Google Scholar] [CrossRef] [Green Version]
  75. Zhang, D.; Tao, X.; Chen, R.; Liu, J.; Li, L.; Fang, X.; Yu, L.; Dai, J. Pericoannosin A, a polyketide synthase−nonribosomal peptide synthetase hybrid metabolite with new carbon skeleton from the endophytic fungus Periconia sp. Org. Lett. 2015, 17, 4304–4307. [Google Scholar] [CrossRef]
  76. Strobel, G.; Yang, X.; Sears, J.; Kramer, R.; Sidhu, R.S.; Hess, W.M. Taxol from Pestalotiopsis microspora, an endophytic fungus of Taxus wallachiana. Microbiology 1996, 142, 435–440. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, J.; Li, G.; Lu, H.; Zheng, Z.; Huang, Y.; Su, W. Taxol from Tubercularia sp. strain TF5, an endophytic fungus of Taxus mairei. FEMS Microbiol. Lett. 2000, 193, 249–253. [Google Scholar] [CrossRef] [Green Version]
  78. Garyali, S.; Kumar, A.; Reddy, M.S. Taxol production by an endophytic fungus, Fusarium redolens, isolated from Himalayan Yew. J. Microbiol. Biotechnol. 2013, 23, 1372–1380. [Google Scholar] [CrossRef] [Green Version]
  79. Kumar, A.; Patil, D.; Rajamohanan, P.R.; Ahmad, A. Isolation, purification and characterization of vinblastine and vincristine from endophytic fungus Fusarium oxysporum isolated from Catharanthus roseus. PLoS ONE 2013, 8, e71805. [Google Scholar] [CrossRef] [Green Version]
  80. Ayob, F.W.; Simarani, K.; Abidin, N.Z.; Mohamed, J. First report on a novel Nigrospora sphaerica isolated from Catharanthus roseus plant with anticarcinogenic properties. Microb. Biotechnol. 2017, 10, 926–932. [Google Scholar] [CrossRef]
  81. Lingqi, Z.; Bo, G.; Haiyan, L.; Songrong, Z.; Hua, S.; Su, G.; Rongcheng, E. Preliminary study on the isolation of endophytic fungus of Catharanthus roseus and its fermentation to produce products of therapeutic value. Chin. Trad. Herb. Drugs 2000, 31, 805–807. [Google Scholar]
  82. Palem, P.C.; Kuriakose, G.C.; Jayabaskaran, C. Endophytic fungus, Talaromyces radicus, isolated from Catharanthus roseus, produces vincristine and vinblastine, which induce apoptotic cell death. PLoS ONE 2015, 11, e0144476. [Google Scholar] [CrossRef] [PubMed]
  83. Kuriakose, G.M.; Palem, P.C.; Jayabaskaran, C. Fungal vincristine from Eutypella spp.—CrP14 isolated from Catharanthus roseus induces apoptosis in human squamous carcinoma cell line-A431. BMC Complem. Altern. Med. 2016, 16, 302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Rehman, S.; Shawl, A.S.; Kour, A.; Andrabi, R.; Sultan, P.; Verma, V.; Qazi, G.N. An endophytic Neurospora sp. from Nothapodytes foetida producing camptothecin. Appl. Biochem. Microb. 2008, 44, 203–209. [Google Scholar] [CrossRef]
  85. Rehman, S.; Shawl, A.S.; Kour, A.; Sultan, P.; Ahmad, K.; Khajuria, R.; Qazi, G.N. Comparative studies and identification of camptothecin produced by an endophyte at shake flask and bioreactor. Nat. Prod. Res. 2009, 23, 1050–1057. [Google Scholar] [CrossRef] [PubMed]
  86. 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. Biot. 2013, 97, 9365–9375. [Google Scholar] [CrossRef] [PubMed]
  87. Ran, X.; Zhang, G.; Li, S.; Wang, J. Characterization and antitumor activity of camptothecin from endophytic fungus Fusarium solani isolated from Camptotheca acuminate. Afr. Health Sci. 2017, 17, 566–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Puri, S.C.; Nazir, A.; Chawla, R.; Arora, R.; Riyaz-ul-Hasan, S.; Amna, T.; Ahmed, B.; Verma, V.; Singh, S.; Sagar, R.; et al. The endophytic fungus Trametes hirsuta as a novel alternative source of podophyllotoxin and related aryl tetralin lignans. J. Biotechnol. 2006, 122, 494–510. [Google Scholar] [CrossRef]
  89. Huang, J.X.; Zhang, J.; Zhang, X.R.; Zhang, K.; Zhang, X.; He, X.R. Mucor fragilis as a novel source of the key pharmaceutical agents podophyllotoxin and kaempferol. Pharm. Biol. 2014, 52, 1237–1243. [Google Scholar] [CrossRef] [PubMed]
  90. Zhu, D.; Wang, J.; Zeng, Q.; Zhang, Z.; Yan, R. A novel endophytic Huperzine A–producing fungus, Shiraia sp. Slf14, isolated from Huperzia serrate. J. Appl. Microbiol. 2010, 109, 1469–1478. [Google Scholar] [CrossRef] [PubMed]
  91. Zhang, Z.B.; Zeng, Q.G.; Yan, R.W.; Wang, Y.; Zou, Z.R.; Zhu, D. Endophytic fungus Cladosporium cladosporioides LF70 from Huperzia serrata produces Huperzine A. World J. Microbiol. Biotechnol. 2011, 27, 479–486. [Google Scholar] [CrossRef]
  92. Sun, J.Q.; Yang, M.H. Huperzine A production by Paecilomyces tenuis YS-13, an endophytic fungus isolated from Huperzia serrata. Nat. Prod. Res. 2015, 29, 1035–1041. [Google Scholar]
  93. Dong, L.H.; Fan, S.W.; Ling, Q.Z.; Huang, B.B.; Wei, Z.J. Indentification of huperzine A–producing endophytic fungi isolated from Huperzia serrate. World J. Microb. Biot. 2014, 30, 1011–1017. [Google Scholar] [CrossRef] [PubMed]
  94. Shi, J.; Zeng, Q.; Liu, Y.; Pan, Z. Alternaria sp. MG1, a resveratrol-producing fungus: Isolation, identification, and optimal cultivation conditions for resveratrol production. Appl. Microbiol. Biotechnol. 2012, 95, 369–379. [Google Scholar] [CrossRef]
  95. Haq, A.; Sofi, N.Y. Vitamin D and breast cancer: Indian perspective. Clin. Nutr. 2017, 12, 1–10. [Google Scholar] [CrossRef] [Green Version]
  96. Iqbal, J.; Abbasi, B.A.; Mahmood, T.; Kanwal, S.; Ali, B.; Khalil, A.T. Plant-derived anticancer agents: A green anticancer approach. Asian Pac. J. Trop. Med. 2017, 7, 1129–1150. [Google Scholar] [CrossRef]
  97. Rice, B.A.; Hoeve, E.S.V.; DeLuca, A.N.; Esserman, L.J.; Rugo, H.P.; Melisko, M.E. Registry study to assess hair loss prevention with the Penguin Cold Cap in breast cancer patients receiving chemotherapy. Breast Cancer Res. Treat. 2018, 167, 117–122. [Google Scholar] [CrossRef]
  98. Zaden, S.Y.B.D.; Qiao, X.; Neefjes, J. New insights into the activities and toxicities of the old anticancer drug doxorubicin. FEBS J. 2020, 288, 6095–6111. [Google Scholar]
  99. Patil, R.; Patil, M.P.; Maheshwari, V.L. Bioactive secondary metabolites from endophytic fungi: A review of biotechnological production and their potential applications. Stud. Nat. Prod. Chem. 2016, 49, 189–205. [Google Scholar]
  100. Uzma, F.; Mohan, C.D.; Hashem, A.; Konappa, N.M.; Rangappa, S.; Kamath, P.V.; Singh, B.P.; Mudili, V.; Gupta, V.K.; Siddaiah, C.N.; et al. Endophytic fungi-alternative source of cytotoxic compounds: A review. Front. Pharmacol. 2018, 9, 309. [Google Scholar] [CrossRef]
  101. Zhao, J.; Zhou, L.; Wang, J.; Shan, T.; Zhong, L.; Liu, X.; Gao, X. Endophytic fungi for producing bioactive compounds originally from their host plants. Curr. Res. Technol. Educ. Trop. Appl. Microbial. Microbial. Biotechnol. 2010, 1, 567–576. [Google Scholar]
  102. Cragg, G.M.; Pezzuto, J.M. Natural products as a vital source for the discovery of cancer chemotherapeutic and chemopreventive agents. Med. Princ. Pract. 2016, 25, 41–59. [Google Scholar] [CrossRef] [PubMed]
  103. Kusari, S.; Lamshoft, M.; Spiteller, M. Aspergillus fumigatus Fresenius, an endophytic fungus from Juniperus communis L. Horstmann as a novel source of the anticancer pro-drug deoxypodophyllotoxin. J. Appl. Microbiol. 2009, 107, 1019–1030. [Google Scholar] [CrossRef]
  104. Mohinudeen, I.A.H.K.; Kanumuri, R.; Soujanya, K.N.; Shaanker, R.U.; Rayala, S.K.; Srivastava, S. Sustainable production of camptothecin from an Alternaria sp. isolated from Nothapodytes nimmoniana. Sci. Rep. 2021, 11, 1478. [Google Scholar] [CrossRef]
  105. Liang, Z.; Xhang, J.; Zhang, X.; Li, J.; Zhanh, X.; Zhao, C. Endophytic fungus from Sinopodophyllum emodi (wall.) ying that produces podophyllotoxin. J. Chromatogr. Sci. 2016, 54, 175–178. [Google Scholar]
  106. Arora, D.; Sharma, N.; Singamaneni, V.; Sharma, V.; Kushwaha, M.; Abrol, V.; Guru, S.; Sharma, S.; Gupta, A.P.; Bhushan, S.; et al. Isolation and characterization of bioactive metabolites from Xylaria psidii, an endophytic fungus of the medicinal plant Aegle marmelos and their role in mitochondrial dependent apoptosis against pancreatic cancer cells. Phytomedicine 2016, 23, 1312–1320. [Google Scholar] [CrossRef]
  107. Li, H.; Xiao, J.; Gao, Y.Q.; Tang, J.J.; Zhang, A.N.; Gao, J.M. Chaetoglobosins from Chaetomium globosum, an endophytic fungus in Ginkgo biloba, and their phytotoxic and cytotoxic activities. J. Agric. Food Chem. 2014, 62, 3734–3741. [Google Scholar] [CrossRef]
  108. Ashoka, G.B.; Shivanna, M.B. Metabolite profiling, in vitro and in silico assessment of antibacterial and anticancer activities of Alternaria alternata endophytic in Jatropha heynei. Arch. Microbiol. 2022, 205, 61. [Google Scholar] [CrossRef]
  109. Kalimuthu, A.K.; Pavadai, P.; Panneerselvam, T.; Babkiewicz, E.; Pijanowska, J.; Mrowka, P.; Rajagopal, G.; Deepak, V.; Sundar, K.; Maszczyk, P.; et al. Cytotoxic potential of bioactive compounds from Aspergillus flavus, an endophytic fungus isolated from Cynodon dactylon, against breast cancer: Experimental and computational approach. Molecules 2022, 27, 8814. [Google Scholar] [CrossRef] [PubMed]
  110. Danagoudar, A.; Joshi, C.G.; Ravi, S.K.; Kumar, H.G.R.; Ramesh, B.N. Antioxidant and cytotoxic potential of endophytic fungi isolated from medicinal plant Tragia involucrata L. Pharmacogn. Res. 2021, 10, 188–195. [Google Scholar]
  111. He, Q.; Zeng, Q.; Shao, Y.; Zhou, H.; Li, T.; Song, F.; Liu, W. Anti-cervical cancer activity of secondary metabolites of endophytic fungi from Ginkgo biloba. Cancer Biomark. 2020, 28, 371–379. [Google Scholar] [CrossRef]
  112. Anoumedem, E.G.M.; Mountessou, B.Y.G.; Kouam, S.F.; Narmani, A.; Surup, F. Simplicilones A and B isolated from the endophytic fungus Simplicillium subtropicum SPC3. Antibiotics 2020, 9, 753. [Google Scholar] [CrossRef]
  113. Sheeba, H.; Ali, M.S.; Anuradha, V. In-vitro anti-cancer Activity of endophytic fungi Isolated from Ziziphus mauritiana in cervical cancer cell line. Eur. J. Med. Plants 2020, 31, 38–48. [Google Scholar] [CrossRef]
  114. Palanichamy, P.; Kannan, S.; Murugan, D.; Alagusundaram, P.; Marudhamuthu, M. Purification, crystallization and anticancer activity evaluation of the compound alternariol methyl ether from endophytic fungi Alternaria alternata. J. Appl. Microbiol. 2019, 127, 1468–1478. [Google Scholar] [CrossRef]
  115. Ukwatta, K.M.; Lawrence, J.L.; Wijayarathne, C.D. Antimicrobial, anti-cancer, anti-filarial and anti-inflammatory activities of cowabenzophenone a extracted from the endophytic fungus Aspergillus terreus isolated from a mangrove plant Bruguiera gymnorrhyza. Mycology 2020, 11, 297–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Kumar, V.S.; Kumaresan, S.; Tamizh, M.M.; Islam, M.I.H.; Thirugnanasambantham, K. Anticancer potential of NF-κB targeting apoptotic molecule “flavipin” isolated from endophytic Chaetomium globosum. Phytomedicine 2019, 61, 152830. [Google Scholar] [CrossRef]
  117. Arifni, F.R.; Hasan, A.E.Z.; Julistiono, H.; Bermawie, N.; Riyanti, E.I. Anticancer activities of endophytic fungi isolated from soursop leaves (Annona muricata L.) against WiDr cancer cells. Annu. Res. Rev. Biol. 2017, 18, 1–11. [Google Scholar] [CrossRef]
  118. Goutam, J.; Sharma, G.; Tiwari, V.K.; Mishra, A.; Kharwar, R.N.; Ramaraj, V.; Koch, B. Isolation and characterization of “Terrin” an antimicrobial and antitumor compound from endophytic fungus Aspergillus terrus (JAS-2) associated from Achyranthus aspera Varanasi, India. Front. Microbiol. 2017, 8, 1–12. [Google Scholar] [CrossRef] [Green Version]
  119. Jinfeng, E.C.; Rafi, M.I.M.; Hoon, K.C.; Lian, H.K.; Kqueen, C.Y. Analysis of chemical constituents, antimicrobial and anticancer activities of dichloromethane extracts of Sordariomycetes sp. endophytic fungi isolated from Strobilanthes crispus. World J. Microb. Biot. 2016, 33, 5. [Google Scholar] [CrossRef]
  120. Wu, L.S.; Jia, M.; Chen, L.; Zhu, B.; Dong, H.X.; Si, J.P.; Peng, W.; Han, T. Cytotoxic and antifungal constituents isolated from the metabolites of endophytic fungus DO14 from Dendrobium officinale. Molecules 2015, 21, 14. [Google Scholar] [CrossRef] [Green Version]
  121. Wibowo, M.; Prachyawarakorn, V.; Aree, T.; Wiyakrutta, S.; Mahidol, C.; Ruchirawat, S.; Kittakoop, P. Tricyclic and spirobicyclic norsesquiterpenes from the endophytic fungus Pseudolagarobasidium acaciicola. Eur. J. Org. Chem. 2014, 19, 3976–3980. [Google Scholar] [CrossRef]
  122. Suja, M.; Vasuki, S.; Sajitha, N. Anticancer activity of compounds isolated from marine endophytic fungus Aspergillus terrus. World J. Pharm. Pharm. Sci. 2014, 3, 661–672. [Google Scholar]
  123. Lin, T.; Wang, G.; Shan, W.; Zeng, D.; Jiang, R.D.X.; Zhu, D.; Liu, X.; Yang, S.; Chen, H. Myrotheciumones: Bicyclic cytotoxic lactones isolated from an endophytic fungus of Ajuga decumbens. Bioorg. Med. Chem. Lett. 2014, 24, 2504–2507. [Google Scholar] [CrossRef] [PubMed]
  124. Verekar, S.A.; Mishra, P.D.; Sreekumar, E.S.; Deshmukh, S.K.; Fiebig, H.H.; Kelter, G.; Maier, A. Anticancer activity of new depsipeptide compound isolated from an endophytic fungus. J. Antibiot. 2014, 67, 697–701. [Google Scholar] [CrossRef] [PubMed]
  125. Zhao, J.; Li, C.; Wang, W.; Zhao, C.; Luo, M.; Mu, F.; Fu, Y.; Zu, Y.; Yao, M. Hypocrea lixii, novel endophytic fungi producing anticancer agent cajanol, isolated from pigeon pea (Cajanus cajan [L.] Millsp). J. Appl. Microbiol. 2013, 115, 102–113. [Google Scholar] [CrossRef]
  126. Zheng, C.J.; Xu, L.L.; Li, Y.Y.; Han, T.; Zhang, Q.Y.; Ming, Q.L.; Rahman, K.; Qin, L.P. Cytotoxic metabolites from the cultures of endophytic fungi from Panax ginseng. Appl. Microbial. Biot. 2013, 97, 7617–7625. [Google Scholar] [CrossRef] [PubMed]
  127. Shweta, S.; Gurumurthy, B.R.; Ravikanth, G.; Ramanan, U.S.; Shivanna, M.B. Endophytic fungi from Miquelia dentata Bedd., produce the anti-cancer alkaloid, camptothecine. Phytomedicine 2013, 20, 337–342. [Google Scholar] [CrossRef] [PubMed]
  128. Kumara, P.M.; Zuehlke, S.; Priti, V.; Ramesha, B.T.; Shweta, S.; Ravikanth, G.; Vasudeva, R.; Santhoshkumar, T.R.; Spiteller, M.; Shaanker, R.M. Fusarium proliferatum, an endophytic fungus from Dysoxylum binectariferum Hook.f, produces rohitukine, a chromane alkaloid possessing anti-cancer activity. Anton Leeuw 2011, 101, 323–329. [Google Scholar] [CrossRef]
  129. Behl, T.; Kaur, I.; Kotwani, A. Implication of oxidative stress in progression of diabetic retinopathy. Surv. Ophthalmol. 2015, 61, 187–196. [Google Scholar] [CrossRef] [PubMed]
  130. Thiruvengadam, M.; Venkidasamy, B.; Subramanian, U.; Samynathan, R.; Shariati, M.A.; Rebezov, M.; Girish, S.; Thangavel, S.; Dhanapal, A.R.; Fedoseeva, N.; et al. Bioactive compounds in oxidative stress-mediated diseases: Targeting the NRF2/ARE signaling pathway and epigenetic regulation. Antioxidants 2021, 10, 1859. [Google Scholar] [CrossRef]
  131. Saxena, P.; Selvaraj, K.; Khare, S.K.; Chaudhary, N. Superoxide dismutase as multipotent therapeutic antioxidant enzyme: Role in human diseases. Biotechnol. Lett. 2021, 44, 1–22. [Google Scholar] [CrossRef] [PubMed]
  132. Gulchin, I. Antioxidants and antioxidant methods: An updated overview. Arch. Toxicol. 2020, 94, 651–715. [Google Scholar] [CrossRef] [Green Version]
  133. Mishra, R.C.; Goel, M.; Barrow, C.J.; Deshmukh, S.K. Endophytic fungi- An untapped source of potential antioxidants. Curr. Bioact. Compd. 2020, 16, 944–964. [Google Scholar] [CrossRef]
  134. Elkhouly, H.I.; Hamed, A.A.; Hosainy, A.M.E.; Ghareeb, M.A.; Sidkey, N.M. Bioactive secondary metabolite from endophytic Aspergillus Tubenginses ASH4 isolated from Hyoscyamus muticus: Antimicrobial, Antibiofilm, Antioxidant and Anticancer Activity. Pharmacogn. J. 2021, 13, 434–442. [Google Scholar] [CrossRef]
  135. Atiphasaworn, P.; Monggoot, S.; Gentekaki, E.; Brooks, S.; Pripdeevech, P. Antibacterial and antioxidant constituents of extracts of endophytic fungi isolated from Ocimum basilicum var. thyrsiflora leaves. Curr. Microbiol. 2017, 74, 1185–1193. [Google Scholar] [CrossRef]
  136. Chatterjee, S.; Ghosh, R.; Mandal, N.C. Production of bioactive compounds with bactericidal and antioxidant potential by endophytic fungus Alternaria alternata AE1 isolated from Azadirachta indica A. Juss. PLoS ONE 2019, 14, e0214744. [Google Scholar] [CrossRef] [Green Version]
  137. Kumaresan, S.; Karthi, V.; Senthilkuma, V.; Balakumar, B.S.; Balakumar, A. Biochemical constituents and antioxidant 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]
  138. Praptiwi, R.M.; Wulansari, D.; Fathoni, A.; Agusta, A. Antibacterial and antioxidant activities of endophytic fungi extracts of medicinal plants from Central Sulawesi. J. Appl. Pharm. Sci. 2018, 8, 69–74. [Google Scholar]
  139. Sadrati, N.; Daoud, H.; Zerroug, A.; Dahamna, S.; Bouharati, S. Screening of antimicrobial and antioxidant secondary metabolites from endophytic fungi isolated from wheat (Triticum durum). J. Plant Prot. Res. 2013, 53, 128–136. [Google Scholar] [CrossRef]
  140. Yadav, M.; Yadav, A.; Yadav, J.P. In vitro antioxidant activity and total phenolic content of endophytic fungi isolated from Eugenia jambolana Lam. Asian Pac. J. Trop. Med. 2014, 7, 256–261. [Google Scholar] [CrossRef] [Green Version]
  141. Nagda, V.; Gajbhiye, A.; Kumar, D. Isolation and characterization of endophytic fungi from Calotropis Procera for their antioxidant activity. Asian J. Pharm. Clin. Res. 2016, 10, 254–258. [Google Scholar] [CrossRef] [Green Version]
  142. Gunasekaran, S.; Sathiavelu, M.; Arunachalam, S. In vitro antioxidant and antibacterial activity of endophytic fungi isolated from Mussaenda luteola. J. Pharm. Sci. 2017, 7, 234–238. [Google Scholar]
  143. Ibrahim, M.; Oyebanji, E.; Fowora, M.; Aiyeolemi, A.; Orabuchi, C.; Akinnawo, B.; Adekunle, A.A. Extracts of endophytic fungi from leaves of selected Nigerian ethnomedicinal plants exhibited antioxidant activity. BMC Complem. Altern. Med. 2021, 21, 98. [Google Scholar] [CrossRef]
  144. Li, Y.L.; Xin, X.M.; Chang, Z.Y.; Shi, R.J.; Miao, Z.M.; Ding, J.; Hao, G.P. The endophytic fungi of Salvia miltiorrhiza Bge.f. alba are a potential source of natural antioxidants. Bot. Stud. 2015, 56, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Selim, K.A.; Elkhateeb, W.A.; Tawila, A.M.; Beih, A.A.E.; Rahman, T.M.A.; Diwany, A.I.E.; Ahmed, E.F. Antiviral and antioxidant potential of fungal endophytes of Egyptian medicinal plants. Fermentation 2018, 4, 49. [Google Scholar] [CrossRef] [Green Version]
  146. Lim, S.M.; Agatonovic-Kustrin, S.A.; Lim, F.T.; Ramasamy, K. High-performance thin layer chromatography-based phytochemical and bioactivity characterisation of anticancer endophytic fungal extracts derived from marine plants. J. Pharmaceut. Biomed. 2021, 193, 113702. [Google Scholar] [CrossRef] [PubMed]
  147. Sharaf, M.H.; Abdelaziz, A.M.; Kalaba, M.H.; Radwan, A.A.; Hashem, A.H. Antimicrobial, antioxidant, cytotoxic activities and phytochemical analysis of fungal endophytes isolated from Ocimum basilicum. Appl. Biochem. Biotechnol. 2021, 194, 1271–1289. [Google Scholar] [CrossRef]
  148. Silva, M.H.R.D.; Yesquen, L.G.C.; Junior, S.B.; Garcia, V.L.; Sartoratto, A.; Angelis, D.D.F.D.; Angelis, D.A.D. Endophytic fungi from Passifora incarnata: An antioxidant compound source. Arch. Microbiol. 2020, 202, 2779–2789. [Google Scholar] [CrossRef]
  149. Tang, Z.; Wang, Y.; Yang, J.; Xiao, Y.; Cai, Y.; Wan, Y.; Chen, H.; Yao, H.; Shan, Z.; Li, C.; et al. Isolation and identification of flavonoid producing endophytic fungi from medicinal plant Conyza blinii H.Lév that exhibit higher antioxidant and antibacterial activities. PeerJ 2020, 8, e8978. [Google Scholar] [CrossRef] [Green Version]
  150. Tian, J.; Fu, L.; Zhang, Z.; Dong, X.; Xu, D.; Mao, Z.; Liu, Y.; Lai, D.; Zhou, L. Dibenzo-α-pyrones from the endophytic fungus Alternaria sp. Samif01: Isolation, structure elucidation, and their antibacterial and antioxidant activities. Nat. Prod. Res. 2017, 31, 387–396. [Google Scholar] [CrossRef]
  151. Prihantini, A.I.; Tachibana, S. Antioxidant compounds produced by Pseudocercospora sp. ESL 02, an endophytic fungus isolated from Elaeocarpus sylvestris. Asian Pac. J. Trop. Biomed. 2016, 7, 110–115. [Google Scholar] [CrossRef]
  152. Dzoyem, J.P.; Melong, R.; Tsamo, A.T.; Maffo, T.; Kapche, D.G.W.F.; Ngadjui, B.T.; Mcgaw, L.J.; Eloff, J.N. Cytotoxicity, antioxidant and antibacterial activity of four compounds produced by an endophytic fungus Epicoccum nigrum associated with Entada abyssinica. Rev. Bras. Farmacogn. 2016, 27, 251–253. [Google Scholar] [CrossRef]
  153. Zhao, J.T.; Ma, D.H.; Luo, M.; Wang, W.; Zhao, C.; Zu, Y.G.; Fu, Y.; Wink, M. In vitro antioxidant activities and antioxidant enzyme activities in HepG2 cells and main active compounds of endophytic fungus from pigeon pea [Cajanus cajan (L.) Millsp.]. Food Res. Int. 2013, 56, 243–251. [Google Scholar] [CrossRef]
  154. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-association diseases in organs. Oncotarget 2017, 9, 7204–7218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Ju, Z.R.; Qin, X.; Lin, X.P.; Wang, J.F.; Kaliyaperumal, K.; Tian, Y.Q.; Liu, J.; Liu, F.; Tu, Z.; Xu, S.H.; et al. New phenyl derivatives from endophytic fungus Botryosphaeria sp. SCSIO KcF6 derived of mangrove plant Kandelia candel. Nat. Prod. Res. 2015, 30, 192–198. [Google Scholar] [CrossRef]
  156. Zhang, M.; Zhao, J.L.; Liu, J.M.; Chen, R.D.; Xie, K.B.; Chen, D.W.; Feng, K.P.; Zhang, D.; Dai, J.G. Neural anti-inflammatory sesquiterpenoids from the endophytic fungus Trichoderma sp. Xy24. J. Asian Nat. Prod. Res. 2016, 19, 651–658. [Google Scholar] [CrossRef]
  157. Harun, A.; Vidyadaran, S.; Lim, S.M.; Cole, A.L.J.; Ramasamy, K. Malaysian endophytic fungal extracts-induced anti-inflammation in Lipopolysaccharide-activated BV-2 microglia is associated with attenuation of NO production and, IL-6 and TNF-α expression. BMC Complem. Altern. Med. 2015, 15, 166. [Google Scholar] [CrossRef] [Green Version]
  158. Mishra, P.D.; Verekar, S.A.; Almeida, A.K.; Roy, S.K.; Jain, S.; Balakrishnan, A.; Vishwakarma, R.; Deshmukh, S.K. Anti-inflammatory and anti-diabetic napthaquinones from an endophytic fungus Dendryphion nanum (Nees) S. Hughes. Indian J. Chem. 2013, 252B, 565–567. [Google Scholar]
  159. Govindappa, M.; Farheen, H.; Chandrappa, C.P.; Rai, R.V.; Raghavendra, V.B. Mycosynthesis of silver nanoparticles using extract of endophytic fungi, Penicillium species of Glycosmis mauritiana, and its antioxidant, antimicrobial, anti-inflammatory and tyrokinase inhibitory activity. Adv. Nat. Sci. Nanosci. Nanotechnol. 2016, 7, 035014. [Google Scholar] [CrossRef]
  160. Moharram, A.M.; Zohri, A.A.; Omar, H.M.; Ghani, O.A.A.E. In vitro assessment of antimicrobial and anti-inflammatory potential of endophytic fungal metabolites extracts. Eur. J. Biol. Res. 2017, 7, 234–244. [Google Scholar]
  161. Chen, Y.; Zou, G.; Yang, W.; Zhao, Y.; Tan, Q.; Chen, L.; Wang, J.; Ma, C.; Kang, W.; She, Z. Metabolites with anti-inflammatory activity from the mangrove endophytic fungus Diaporthe sp. QYM12. Mar. Drugs 2021, 19, 56. [Google Scholar] [CrossRef] [PubMed]
  162. Liu, H.; Yan, C.; Li, C.; You, T.; She, Z. Naphthoquinone derivatives with anti-inflammatory activity from mangrove-derived endophytic fungus Talaromyces sp. SK-S009. Molecules 2020, 25, 576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Tan, Y.; Guo, Z.; Zhu, M.; Shi, J.; Li, W.; Jiao, R.; Tan, R.; Ge, H. Anti-inflammatory spirobisnaphthalene natural products from a plant-derived endophytic fungus Edenia gomezpompae. Chin. Chem. Lett. 2020, 31, 1406–1409. [Google Scholar] [CrossRef]
  164. Chen, Y.; Zhang, L.; Zou, G.; Li, C.; Yang, W.; Liu, H.; She, Z. Anti-inflammatory activities of alkaloids from the mangrove endophytic fungus Phomopsis sp. SYSUQYP-23. Bioorg. Chem. 2020, 97, 103712. [Google Scholar] [CrossRef]
  165. Rabia, M.W.A.; Mohamed, G.A.; Ibrahim, S.R.M.; Asfour, H.Z. Anti-inflammatory ergosterol derivatives from the endophytic fungus Fusarium chlamydosporum. Nat. Prod. Res. 2020, 35, 5011–5020. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, N.N.; Liu, C.Y.; Wang, T.; Li, Y.L.; Xu, K.; Lou, H.X. Two new quinazoline derivatives from the moss endophytic fungus Aspergillus sp. and their antiinfammatory activity. Nat. Prod. Bioprospect. 2021, 11, 105–110. [Google Scholar] [CrossRef]
  167. Khayat, M.T.; Ibrahim, S.R.M.; Mohamed, G.A.; Abdallah, H.M. Anti-inflammatory metabolites from endophytic fungus Fusarium sp. Phytochem. Lett. 2019, 29, 104–109. [Google Scholar] [CrossRef]
  168. Liao, G.; Wu, P.; Xue, J.; Liu, L.; Li, H.; Wei, X. Asperimides A–D, anti-inflammatory aromatic butenolides from a tropical endophytic fungus Aspergillus terreus. Fitoterapia 2018, 131, 50–54. [Google Scholar] [CrossRef] [PubMed]
  169. Chen, Y.; Liu, Z.; Liu, H.; Pan, Y.; Li, J.; Liu, L.; She, Z. Dichloroisocoumarins with potential anti-inflammatory activity from the mangrove endophytic fungus Ascomycota sp. CYSK-4. Mar. Drugs 2018, 16, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Chen, S.; Liu, Z.; Liu, H.; Long, Y.; Chen, D.; Lu, Y.; She, Z. Lasiodiplactone A, a novel lactone from the mangrove endophytic fungus Lasiodiplodia theobromae ZJ-HQ1. Org. Biomol. Chem. 2017, 15, 6338–6341. [Google Scholar] [CrossRef] [PubMed]
  171. Guo, F.; Li, Z.; Xu, X.; Wang, K.; Shao, M.; Zhao, F.; Wang, H.; Hua, H.; Pei, Y.; Bai, J. Butenolide derivatives from the plant endophytic fungus Aspergillus terreus. Fitoterapia 2016, 113, 44–50. [Google Scholar] [CrossRef] [PubMed]
  172. Zhang, D.; Ge, H.; Zou, J.H.; Tao, X.; Chen, R.; Dai, J. Periconianone A, a new 6/6/6 carbocyclic sesquiterpenoid from endophytic fungus Periconia sp. with neural anti-inflammatory activity. Org. Lett. 2014, 16, 1410–1413. [Google Scholar] [CrossRef] [PubMed]
  173. Uzor, P.F.; Osadebe, P.O.; Nwodo, N.J. Antidiabetic activity of extract and compounds from an endophytic fungus Nigrospora oryzae. Drug Res. 2016, 67, 308–311. [Google Scholar] [CrossRef]
  174. Rivera-Chavez, J.; Gonzalez-Andrade, M.; Gonzalez, M.D.C.; Glenn, A.E.; Mata, R. Thielavins A, J and K: A-Glucosidase inhibitors from MEXU 27095, an endophytic fungus from Hintonia latiflora. Phytochemistry 2013, 94, 198–205. [Google Scholar] [CrossRef]
  175. Ye, G.; Huang, C.; Li, J.; Chen, T.; Tang, J.; Liu, W.; Long, Y. Isolation, structural characterization and antidiabetic activity of new diketopiperazine alkaloids from mangrove endophytic fungus Aspergillus sp. 16-5c. Mar. Drugs 2021, 19, 402. [Google Scholar] [CrossRef]
  176. Singh, B.; Kaur, A. Antidiabetic potential of a peptide isolated from an endophytic Aspergillus awamori. J. Appl. Microbiol. 2015, 120, 301–311. [Google Scholar] [CrossRef] [Green Version]
  177. Ruzieva, D.M.; Abdulmyanova, L.I.; Rasulova, G.A.; Sattarova, R.S.; Gulyamova, T.G. Screening of inhibitory activity against α-amylase of fungal endophytes isolated from medicinal plants in Uzbekistan. Int. J. Curr. Microbiol. Appl. Sci. 2017, 6, 2744–2752. [Google Scholar]
  178. Ruzieva, D.; Gulyamova, T.; Nasmetova, S.; Mukhammedov, I.; Rasulova, G. Identification of Bioactive Compounds of the Endophytic Fungus Aspergillus egypticus-HT166S Inhibiting the Activity of Pancreatic α-Amylase. Turk. J. Pharm. Sci. 2022, 19, 630–635. [Google Scholar] [CrossRef]
  179. Pavithra, N.; Sathish, L.; Babu, N.; Venkatarathanamma, V.; Pushpalatha, H.; Reddy, G.B.; Ananda, K. Evaluation of α-Amylase, α-glucosidase and aldose reductase inhibitors in ethyl acetate extracts of endophytic fungi isolated from antidiabetic medicinal plants. Int. J. Pharm. Sci. Res. 2014, 5, 5334–5341. [Google Scholar]
  180. Ali, S.; Khan, A.L.; Ali, L.; Rizvi, T.S.; Khan, S.A.; Hussain, J.; Hamayun, M.; Al-Harrasi, A. Enzyme inhibitory metabolites from endophytic Penicillium citrinum isolated from Boswellia sacra. Arch. Microbiol. 2017, 199, 691–700. [Google Scholar] [CrossRef] [PubMed]
  181. Song, Y.; Wang, J.; Huang, H.; Ma, L.; Wang, J.; Gu, Y.; Liu, L.; Lin, Y. Four Eremophilane Sesquiterpenes from the Mangrove Endophytic Fungus Xylaria sp. Mar. Drugs 2012, 10, 340–348. [Google Scholar] [CrossRef] [Green Version]
  182. Xia, X.; Qi, J.; Liu, I.; Jia, A.; Zhang, Y.; Liu, C.; Gao, C.; She, Z. Bioactive isopimarane diterpenes from the fungus, Epicoccum sp. HS-1 associated with Apostichopus japonicas. Mar. Drugs 2015, 13, 1124–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Lopéz, D.; Cherigo, L.; Mejia, L.C.; Loza-Mejía, M.A.; Martínez-Luis, S. α-Glucosidase inhibitors from a mangrove associated fungus, Zasmidium sp. strain EM5-10. BMC Chem. 2019, 13, 22. [Google Scholar] [CrossRef] [Green Version]
  184. Malik, A.; Ardalani, H.; Anam, S.; McNair, L.M.; Kromphardt, K.J.K.; Frandsen, R.J.N.; Franzyk, H.; Staerk, D.; Kongstad, K.T. Antidiabetic xanthones with α-glucosidase inhibitory activities from an endophytic Penicillium canescens. Fitoterapia 2020, 142, 104522. [Google Scholar] [CrossRef]
  185. Greener, M. Autoimmune diseases: The immunological tightrope. Presecriber 2022, 33, 19–22. [Google Scholar] [CrossRef]
  186. Mohammedsaleh, Z.M. The use of patient-specific stem cells in different autoimmune diseases. Saudi J. Biol. Sci. 2022, 29, 3338–3346. [Google Scholar] [CrossRef] [PubMed]
  187. Shukla, S.T.; Habbu, P.V.; Kulkarni, V.H.; Jagadish, S.K.; Pandey, A.R.; Sutariya, V.N. Endophytic microbes: A novel source for biologically/pharmacologically active secondary metabolites. Asian J. Pharmacol. Toxicol. 2014, 2, 1–16. [Google Scholar]
  188. Ujam, N.T.; Ajaghaku, D.L.; Okoye, F.B.C.; Esimone, C.O. Antioxidant and immunosuppressive activities of extracts of endophytic fungi isolated from Psidium guajava and Newbouldia laevis. Phytomed. Plus 2021, 1, 100028. [Google Scholar] [CrossRef]
  189. Wang, L.W.; Wang, J.L.; Chen, J.; Chen, J.J.; Shen, J.W.; Feng, X.X.; Kubicek, C.P.; Lin, F.C.; Zhang, C.L.; Chen, F.Y. A novel derivative of (-) mycousnine produced by the endophytic fungus Mycosphaerella nawae, exhibits high and selective immunosuppressive activity on T cells. Front. Microbiol. 2017, 8, 1251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Liu, H.; Chen, S.; Liu, W.; Liu, Y.; Huang, X.; She, Z. Polyketides with immunosuppressive activities from mangrove endophytic fungus Penicillium sp. ZJ-SY2. Mar. Drugs 2016, 14, 217. [Google Scholar] [CrossRef] [Green Version]
  191. Lin, R.; Kim, H.; Hong, J.; Li, Q.J. Biological evaluation of subglutinol a as a novel immunosuppressive agent for inflammation intervention. Med. Chem. Lett. 2014, 5, 485–490. [Google Scholar] [CrossRef]
  192. Wei, W.; Gao, J.; Shen, Y.; Chu, Y.L.; Xu, Q.; Tan, R.X. Immunosuppressive Diterpenes from Phomopsis sp. S12. Eur. J. Org. Chem. 2014, 26, 5728–5734. [Google Scholar] [CrossRef]
  193. Katoch, M.; Khajuria, A.; Sharma, P.R.; Saxena, A.K. Immunosuppressive potential of Botryosphaeria dothidea, an endophyte isolated from Kigelia Africana. Pharm. Biol. 2014, 53, 85–91. [Google Scholar] [CrossRef]
  194. Ye, K.; Lv, X.; Zhang, X.; Wei, P.P.; Li, Z.H.; Ai, H.L.; Zhao, D.K.; Liu, J.K. Immunosuppressive isopimarane diterpenes from cultures of the endophytic fungus Ilyonectria robusta. Front. Pharmacol. 2022, 12, 3847. [Google Scholar] [CrossRef]
  195. Lin, S.; Yan, S.; Liu, Y.; Zhang, X.; Cao, F.; He, Y.; Li, F.; Liu, J.; Wang, J.; Hu, J.; et al. New secondary metabolites with immunosuppressive and BChE inhibitory activities from an endophytic fungus Daldinia sp. TJ403-LS1. Bioorg. Chem. 2021, 114, 105091. [Google Scholar] [CrossRef]
  196. Zhang, W.F.; Ma, J.K.; Zhang, X.X.; Qian, Y.N.; Xu, J. Immunosuppressive polyketides from the mangrove endophytic fungus Pestalotiopsis sp. HHL-14. Chem. Nat. Compd. 2021, 57, 1130–1133. [Google Scholar] [CrossRef]
  197. Xu, Z.Y.; Zhang, X.X.; Ma, J.K.; Yang, Y.; Zhou, J.; Xu, J. Secondary metabolites produced by mangrove endophytic fungus Aspergillus fumigatus HQD24 with immunosuppressive activity. Bochem. Syst. Ecol. 2020, 93, 104166. [Google Scholar] [CrossRef]
  198. Sandhu, S.S.; Kumar, S.; Aharwal, R.P. Isolation and identification of endophytic fungi from Ricinus communis Linn. and their antibacterial activity. Int. J. Pharm. Pharm. Sci. 2014, 4, 611–618. [Google Scholar]
  199. Mc-Mullin, D.R.; Green, B.D.; Miller, J.D. Antifungal sesquterpenoids and macrolides from an endophytic Lophodernium species of Pinus strobus. Phytochem. Lett. 2015, 14, 148–152. [Google Scholar] [CrossRef]
  200. Zhou, M.; Zhou, K.; He, P.; Wang, K.M.; Zhu, R.Z.; Wang, Y.D.; Dong, W.; Li, G.P.; Yang, H.Y.; Ye, Y.Q.; et al. Antiviral and cytotoxic isocoumarin derivatives from an endophytic fungus Aspergillus oryzae. Planta Med. 2015, 82, 414–417. [Google Scholar] [CrossRef] [Green Version]
  201. Mishra, V.K.; Passari, A.K.; Chandra, P.; Leo, V.V.; Kumar, B.; Uthandi, S.; Thankappan, S.; Gupta, V.K.; Singh, B.P. Determination and production of antimicrobial compounds by Aspergillus clavatonanicus strain MJ31, an endophytic fungus from Mirabilis jalapa L. using UPLC-ESIMS/MS and TD-GC-MS analysis. PLoS ONE 2017, 12, e0186234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Guo, L.; Niu, S.; Chen, S.; Liu, L. Diaporone A, a new antibacterial secondary metabolite from the plant endophytic fungus Diaporthe sp. J. Antibiot. 2019, 73, 116–119. [Google Scholar] [CrossRef] [PubMed]
  203. Yehia, R.; Shivanna, M.B. Antimicrobial activity and metabolite profling of endophytic fungi in Digitaria bicornis (Lam) Roem. and Schult. and Paspalidium favidum (Retz.) A. Camus. 3 Biotech 2021, 11, 53. [Google Scholar]
  204. Zihad, S.M.N.K.; Hasan, M.T.; Sultana, M.S.; Nath, S.; Nahar, L.; Rashid, M.A.; Uddin, S.J.; Sarker, S.D.; Shilpi, J.A. Isolation and characterization of antibacterial compounds from Aspergillus fumigatus: An endophytic fungus from a mangrove plant of the Sundarbans. Hindawi Evi. Based Complement. Altern. Med. 2022, 2022, 9600079. [Google Scholar] [CrossRef] [PubMed]
  205. Elkady, W.M.; Raafat, M.M.; Abdel-Aziz, M.M.; Al-Huqail, A.A.; Ashour, M.L.; Fathallah, N. Endophytic fungus from Opuntina fiscus-indica: A source of potential bioactive antimicrobial compounds against multidrug-resistant bacteria. Plants 2022, 11, 1070. [Google Scholar] [CrossRef]
  206. Xian, P.J.; Liu, S.Z.; Wang, W.J.; Yang, S.X.; Feng, Z.; Yang, X.L. Undescribed specialised metabolites from the endophytic fungus Emericella sp. XL029 and their antimicrobial activities. Phytochemistry 2022, 202, 113303. [Google Scholar] [CrossRef]
  207. Astuti, P.; Rollando, R.; Pratoko, D.K.; Wahyuono, S.; Nurrochmad, A. Antimicrobial sand cytotoxic activities of a compound produced by an endophytic fungus isolated from the leaves of Coleus amboinicus Lou. Int. J. Pharm. Res. 2020, 13, 2632–2644. [Google Scholar]
  208. Khiralla, A.; Spina, R.; Varbanov, M.; Philippot, S.; Lemiere, P.; Deschaumes, S.S.; Andre, P.P.; Mohamed, I.; Yagi, S.M.; Mattar, D.L. Evaluation of antiviral, antibacterial and antiproliferative activities of the endophytic fungus Curvularia papendorfii, and isolation of a new polyhydroxyacid. Microorganisms 2020, 8, 1353. [Google Scholar] [CrossRef]
  209. Techaoei, S.; Jirayuthcharoenkul, C.; Jarmkom, K.; Dumrongphuttidecha, T.; Khobjai, W. Chemical evaluation and antibacterial activity of novel bioactive compounds from endophytic fungi in Nelumbo nucifera. Saudi J. Biol. Sci. 2020, 27, 2883–2889. [Google Scholar] [CrossRef]
  210. Hilario, F.; Polinario, G.; Amorim, M.R.D.; Batista, V.D.S.; Junior, N.M.D.N.; Araujo, A.R.; Bauab, T.R.; Santos, L.C.D. Spirocyclic lactams and curvulinic acid derivatives from the endophytic fungus Curvularia lunata and their antibacterial and antifungal activities. Fitoterapia 2019, 141, 104466. [Google Scholar] [CrossRef] [PubMed]
  211. Zhang, P.; Yuan, X.L.; Du, Y.M.; Zhang, H.B.; Shen, G.M.; Zhang, Z.F.; Liang, Y.J.; Zhao, D.L.; Xu, K. Angularly prenylated indole alkaloids with antimicrobial and insecticidal activities from an endophytic fungus Fusarium sambucinum TE-6L. J. Agr. Food Chem. 2019, 67, 11994–12001. [Google Scholar] [CrossRef] [PubMed]
  212. Kaaniche, F.; Hamed, A.; Razek, A.S.A.; Wibberg, D.; Abdissa, N.; Euch, I.Z.E.; Allouche, N.; Mellouli, L.; Shaaban, F.; Sewald, N. Bioactive secondary metabolites from new endophytic fungus Curvularia. sp. isolated from Rauwolfia macrophylla. PLoS ONE 2019, 14, e0217627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Pansanit, A.; Pripdeevech, P. Antibacterial secondary metabolites from an endophytic fungus, Arthrinium sp. MFLUCC16-1053 isolated from Zingiber cassumunar. Mycology 2018, 9, 264–272. [Google Scholar] [CrossRef] [Green Version]
  214. Wang, W.X.; Kusari, S.; Laatsch, H.; Golz, C.; Kusari, P.; Strohmann, C.; Kayser, O.; Spiteller, M. Antibacterial Azaphilones from an endophytic fungus, Colletotrichum sp. BS4. J. Nat. Prod. 2016, 79, 704–710. [Google Scholar] [CrossRef] [Green Version]
  215. Chen, S.; Chen, D.; Cai, R.; Cui, H.; Long, Y.; Lu, Y.; Li, C.; She, Z. Cytotoxic and antibacterial preussomerins from the mangrove endophytic fungus Lasiodiplodia theobromae ZJ-HQ1. J. Nat. Prod. 2016, 79, 2397–2402. [Google Scholar] [CrossRef]
  216. Goutam, J.; Kharwar, R.N.; Kharwar, V.K.; Mishra, A.; Singh, S. Isolation and identification of antibacterial compounds isolated from endophytic fungus Emericella qaudrilineata (RS-5). Nat. Prod. Chem. Res. 2016, 4, 2–7. [Google Scholar] [CrossRef]
  217. Meng, L.H.; Zhang, P.; Li, X.M.; Wang, B.G. Penicibrocazines A–E, five new sulfide diketopiperazines from the marine-derived endophytic Fungus Penicillium brocae. Mar. Drugs 2015, 13, 276–287. [Google Scholar] [CrossRef] [Green Version]
  218. Ratnaweera, P.B.; Silva, E.D.D.; Williams, D.E.; Andersen, R.J. Antimicrobial activities of endophytic fungi obtained from the arid zone invasive plant Opuntia dillenii and the isolation of equisetin, from endophytic Fusarium sp. BMC Compl. Altern. Med. 2015, 15, 220. [Google Scholar] [CrossRef] [Green Version]
  219. Hussain, H.; Root, N.; Jabeen, F.; Harrasi, A.A.; Ahmad, M.; Mabood, F.; Hassan, Z.; Shah, A.; Green, I.R.; Schulz, B.; et al. Microsphaerol and seimatorone: Two new compounds isolated from the endophytic fungi, Microsphaeropsis sp. and Seimatosporium sp. Chem. Biodivers. 2014, 12, 289–294. [Google Scholar] [CrossRef] [PubMed]
  220. Hussain, H.; Root, N.; Jabeen, F.; Harrasi, A.A.; Rawahi, A.A.; Ahmad, M.; Hassan, Z.; Abbas, G.; Mabood, F.; Shah, A.; et al. Seimatoric acid and colletonoic acid: Two new compounds from the endophytic fungi, Seimatosporium sp. and Colletotrichum sp. Chin. Chem. Lett. 2014, 25, 1577–1579. [Google Scholar] [CrossRef]
  221. Li, G.; Kusari, S.; Lamshoft, M.; Schuffler, A.; Laatsch, H.; Spiteller, M. Antibacterial secondary metabolites from an endophytic fungus, Eupenicillium sp. LG41. J. Nat. Prod. 2014, 77, 2335–2341. [Google Scholar] [CrossRef]
  222. Ding, X.; Liu, K.; Deng, B.; Chen, W.; Li, W.; Liu, F. Isolation and characterization of endophytic fungi from Camptotheca acuminata. World J. Microb. Biot. 2013, 29, 1831–1838. [Google Scholar] [CrossRef]
  223. Zhang, H.; Xiong, Y.; Zhao, H.; Yi, Y.; Zhang, C.; Yu, C.; Xu, C. An antimicrobial compound from the endophytic fungus Phoma sp. isolated from the medicinal plant Taraxacum mongolicum. J. Taiwan Inst. Chem. E 2012, 44, 177–181. [Google Scholar] [CrossRef]
  224. Zhao, J.; Sun, W.; Shan, T.; Mou, Y.; Lou, J.; Li, Y.; Wang, M.; Zhou, L. Antimicrobial metabolites from the endophytic fungus Gliomastix murorum Ppf8 associated with the medicinal plant Paris polyphylla var. yunnanensis. J. Med. Plants Res. 2012, 6, 2100–2104. [Google Scholar]
  225. Devi, P.; Rodrigues, C.; Naik, C.G.; D’Souza, L. Isolation and characterization of antibacterial compound from a mangrove-endophytic fungus, Penicillium chrysogenum MTCC 5108. Indian J. Microbiol. 2012, 52, 617–623. [Google Scholar] [CrossRef] [Green Version]
  226. Sebastianes, F.L.S.; Cabedo, N.; Aouad, N.E.; Valente, A.M.M.P.; Lacava, P.T.; Azevedo, J.L.; Kleiner, A.A.P.; Cortes, D. 3-Hydroxypropionic acid as an antibacterial agent from endophytic fungi Diaporthe phaseolorum. Curr. Microbiol. 2012, 65, 622–632. [Google Scholar] [CrossRef]
  227. Pinheiro, E.A.A.; Carvalho, J.M.; Santos, D.C.P.D.; Feitosa, A.D.O.; Marinho, P.S.B.; Guilhon, G.M.S.P.; Souza, A.D.L.D.; Silva, F.M.A.D.; Marinho, A.M.D.R. Antibacterial activity of alkaloids produced by endophytic fungus Aspergillus sp. EJC08 isolated from medical plant Bauhinia guianensis. Nat. Prod. Res. 2012, 27, 1633–1638. [Google Scholar] [CrossRef]
  228. Meng, X.; Mao, Z.; Lou, J.; Xu, L.; Zhong, L.; Peng, Y.; Zhou, L.; Wang, M. Benzopyranones from the endophytic fungus Hyalodendriella sp. Ponipodef12 and their bioactivities. Molecules 2012, 17, 11303–11314. [Google Scholar] [CrossRef]
  229. Erbert, C.; Lopes, A.A.; Yokoya, N.S.; Furtado, N.A.J.C.; Conti, R.; Pupo, M.T.; Lopes, J.L.C.; Debonsi, H.M. Antibacterial compound from the endophytic fungus Phomopsis longicolla isolated from the tropical red seaweed Bostrychia radicans. Bot. Mar. 2012, 55, 435–440. [Google Scholar] [CrossRef]
  230. Subban, K.; Subramani, R.; Johnpaul, M. A novel antibacterial and antifungal phenolic compound from the endophytic fungus Pestalotiopsis mangiferae. Nat. Prod. Res. 2012, 27, 1445–1449. [Google Scholar] [CrossRef] [PubMed]
  231. Qin, S.; Krohn, K.; Hussain, H.; Schulz, B.; Draeger, S. Pestalotheols E–H: Antimicrobial metabolites from an endophytic fungus isolated from the tree Arbutus unedo. Eur. J. Org. Chem. 2011, 2011, 5163–5166. [Google Scholar] [CrossRef]
  232. Ahmed, I.; Hussain, H.; Schulz, B.; Draeger, S.; Padula, D.; Pescitelli, G.; Ree, T.V.; Krohn, K. Three new antimicrobial metabolites from the endophytic fungus Phomopsis sp. Eur. J. Org. Chem. 2011, 2011, 2867–2873. [Google Scholar] [CrossRef]
  233. Nithya, K.; Muthumary, J. Bioactive metabolite produced by Phomopsis sp., an endophytic fungus in Allamanda cathartica Linn. Recent Res. Sci. Technol. 2011, 3, 44–48. [Google Scholar]
  234. Yehia, R.S.; Osman, G.H.; Assaggaf, H.; Salem, R. Isolation of potential antimicrobial metabolites from endophytic fungus Cladosporium cladosporioides from endemic plant Zygophyllum mandavillei. S. Afr. J. Bot. 2020, 15, 313–319. [Google Scholar] [CrossRef]
  235. Gao, Y.; Wang, L.; Kalscheuer, R.; Liu, Z.; Proksch, P. Antifungal polyketide derivatives from the endophytic fungus Aplosporella javeedii. Bioorg. Med. Chem. 2020, 28, 115456. [Google Scholar] [CrossRef]
  236. Luo, H.; Qing, Z.; Deng, Y.; Deng, Z.; Tang, X.; Feng, B.; Lin, W. Two polyketides produced by endophytic Penicillium citrinum DBR-9 from medicinal plant Stephania kwangsiensis and their antifungal activity against plant pathogenic fungi. Nat. Prod. Commun. 2019, 14, 1–6. [Google Scholar] [CrossRef] [Green Version]
  237. Bungtongdee, N.; Sopalun, K.; Laosripaiboon, W.; Lamtham, S. The chemical composition, antifungal, antioxidant and antimutagenicity properties of bioactive compounds from fungal endophytes associated with Thai orchids. J. Phytopathol. 2018, 167, 56–64. [Google Scholar] [CrossRef]
  238. Venkateshwarulu, N.; Shameer, S.; Bramhachari, P.V.; Basha, S.K.T.; 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. [Google Scholar] [CrossRef]
  239. Premjanu, N.; Jaynthy, C.; Diviya, S. Antifungal activity of endophytic fungi isolated from Lannea coromandelicaan in-silico approach. Int. J. Pharm. Pharm. Sci. 2016, 8, 207–210. [Google Scholar]
  240. Pereira, C.B.; Oliveira, D.M.J.; Hughes, A.F.S.; Kohlhof, M.; Vieira, M.L.A.; Vaz, A.B.M.; Ferreira, M.C.; Carvalho, C.R.; Rosa, L.H.; Rosa, C.A.; et al. Endophytic fungal compounds active against Cryptococcus neoformans and C. gattii. J. Antibiot. 2015, 68, 436–444. [Google Scholar] [CrossRef] [PubMed]
  241. Nalli, Y.; Mirza, D.N.; WAni, Z.A.; Wadhwa, B.; Mallik, F.A.; Raina, C.; Chaubey, A.; Riyaz-UI-Hassan, S.; Ali, A. Phialomustin A-D, new antimicrobial and cytotoxic metabolites from an endophytic fungus, Phialophora mustea. R. Soc. Chem. 2015, 5, 95307–95312. [Google Scholar] [CrossRef]
  242. Taware, R.; Abnave, P.; Patil, D.; Rajamohananan, P.R.; Raja, R.; Soundararajan, G.; Kundu, G.C.; Ahmad, A. Isolation, purification and characterization of Trichothecinol-A produced by endophytic fungus Trichothecium sp. and its antifungal, anticancer and antimetastatic activities. Sustain Chem. Process 2014, 2, 8. [Google Scholar] [CrossRef] [Green Version]
  243. Sumarah, M.W.; Kesting, J.R.; Sorensen, D.; Miller, J.D. Antifungal metabolites from fungal endophytes of Pinus strobus. Phytochemistry 2011, 72, 1833–1837. [Google Scholar] [CrossRef]
  244. Sun, Z.L.; Zhang, M.; Zhang, J.F.; Feng, F. Antifungal and cytotoxic activities of the secondary metabolites from endophytic fungus Massrison sp. Phytomedicine 2011, 18, 859–862. [Google Scholar] [CrossRef] [PubMed]
  245. EI-Hawary, S.S.; Mohammed, R.; Bahr, H.S.; Attia, E.Z.; EI-Katatny, M.H.; Abelyan, N.; AI-Sanea, M.M.; Moawad, A.S.; Abdelmohsen, U.R. Soyabean-associated endophytic fungi as potential source of anti-COVID-19 metabolites supported by docking analysis. J. Appl. Microbiol. 2021, 131, 1193–1211. [Google Scholar] [CrossRef]
  246. Liu, S.S.; Jiang, J.X.; Huang, R.; Wang, Y.T.; Jiang, B.G.; Zheng, K.X.; Wu, S.H. A new antiviral 14-nordrimane sesquiterpenoid from an endophytic fungus Phoma sp. Phytochem. Lett. 2019, 29, 75–78. [Google Scholar] [CrossRef]
  247. Liang, H.Q.; Zhang, D.W.; Guo, S.X.; Yu, J. Two new tetracyclic triterpenoids from the endophytic fungus Hypoxylon sp. 6269. J. Asian Nat. Prod. Res. 2018, 20, 951–956. [Google Scholar] [CrossRef]
  248. Pang, X.; Zhao, J.Y.; Fang, X.M.; Zhang, T.; Zhang, D.W.; Liu, H.Y.; Su, J.; Cen, S.; Yu, L.Y. Metabolites from the plant endophytic fungus Aspergillus sp. CPCC 400735 and their anti-HIV activities. J. Nat. Prod. 2017, 80, 2595–2601. [Google Scholar] [CrossRef]
  249. Zhang, S.P.; Huang, R.; Li, F.F.; Wei, H.X.; Fang, X.W.; Xie, X.S.; Lin, D.G.; Wu, S.H.; He, J. Antiviral anthraquinones and azaphilones produced by an endophytic fungus Nigrospora sp. from Aconitum carmichaeli. Fitoterapia 2016, 112, 85–89. [Google Scholar] [CrossRef]
  250. Uzor, P.F.; Odimegwu, D.C.; Ebrahim, W.; Osadebe, P.O.; Nwodo, N.J.; Okoye, F.B.; Liu, Z.; Proksch, P. Anti-Respiratory Syncytial Virus Compounds from Two Endophytic Fungi Isolated from Nigerian Medicinal Plants. Drug Res. (Stuttg) 2016, 66, 527–531. [Google Scholar] [CrossRef] [PubMed]
  251. Govindappa, M.; Hemmanur, K.C.; Nithin, S.; Poojari, C.C.; Kakunje, G.B.; Channabasava, K. In vitro anti-HIV activity of partially purified coumarin(s) isolated from fungal endophyte, Alternaria species of Calophyllum inophyllum. Pharm. Pharmacol. 2015, 6, 321–328. [Google Scholar] [CrossRef] [Green Version]
  252. Chen, C.; Zhu, H.; Wang, J.; Yang, J.; Li, X.N.; Wang, J.; Chen, K.; Wang, Y.; Luo, Z.; Yao, G.; et al. Armochaetoglobins K–R, anti-HIV pyrrole-based cytochalasans from Chaetomium globosum TW1-1. Eur. J. Org. Chem. 2015, 13, 3086–3094. [Google Scholar] [CrossRef]
  253. Liu, X.; Guo, W.J.; Fu, W.J.; Fan, M. Steroids from an anti-HIV active endophyte. Chem. Nat. Compd. 2014, 50, 387–388. [Google Scholar] [CrossRef]
  254. Bashyal, B.P.; Wellensiek, B.P.; Ramakrishnan, R.; Faeth, S.H.; Ahmad, N.; Gunatilaka, A.A.L. Altertoxins with potent anti-HIV activity from Alternaria tenuissima QUE1Se, a fungal endophyte of Quercus emoryi. Bioorg. Med. Chem. 2014, 22, 6112–6116. [Google Scholar] [CrossRef] [Green Version]
  255. Wellensiek, B.P.; Ramakrishnan, R.; Bashyal, B.P.; Eason, Y.; Gunatilaka, A.A.L.; Ahmad, N. Inhibition of HIV-1 replication by secondary metabolites from endophytic fungi of desert plants. Virol. J. 2013, 7, 72–80. [Google Scholar] [CrossRef] [Green Version]
  256. Zhang, G.; Sun, S.; Zhu, T.; Gu, Z.L.J.; Li, D.; Gu, Q. Antiviral isoindolone derivatives from an endophytic fungus Emericella sp. associated with Aegiceras corniculatum. Phytochemistry 2011, 72, 1436–1442. [Google Scholar] [CrossRef]
  257. Gendy, M.M.A.E.; Bondkly, A.M.A.E.; Yahya, S.M.M. Production and evaluation of antimycotic and antihepatitis C virus potential of Fusant MERV6270 derived from mangrove endophytic fungi using novel substrates of agroindustrial wastes. Appl. Biochem. Biotech. 2014, 174, 2674–2701. [Google Scholar] [CrossRef]
  258. Cholewinski, M.; Derda, M.; Hadas, E. Parasitic diseases in humans transmitted by vectors. Ann. Parasitol. 2015, 61, 137–157. [Google Scholar]
  259. Capela, R.; Moreira, R.; Lopes, F. An overview of drug resistancein protozoal diseases. Int. J. Mol. Sci. 2019, 20, 5748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Fernandez-Pastor, I.; Gonzalez-Menéndez, V.; Annang, F.; Toro, C.; Mackenzie, T.A.; Bosch-Navarrete, C.; Genilloud, O.; Reyes, F. Pipecolisporin, a novel cyclic peptide with antimalarial and antitrypanosome activities from a wheat endophytic Nigrospora oryzae. Pharmaceuticals 2021, 14, 268. [Google Scholar] [CrossRef]
  261. Alhadrami, H.A.; Sayed, A.M.; El Gendy, A.O.; Shamikh, Y.I.; Gaber, Y.; Bakeer, W.; Sheirf, N.H.; Attia, E.Z.; Shaban, G.M.; Khalifa, B.A.; et al. A metabolomic approach to target antimalarial metabolites in the Artemisia annua fungal endophytes. Sci. Rep. 2021, 11, 2770. [Google Scholar] [CrossRef]
  262. Purwantini, I.; Wahyono, M.; Susidarti, R.A.; Sholikhah, E.N.; Hestiyani, R.A.N. Antiplasmodial activity of endophytic fungi isolated from Artemisia Annua, L. Int. J. Pharm. Clin. Res. 2016, 8, 341–344. [Google Scholar]
  263. Kumarihamy, M.; Ferreira, D.; Croom, E.C.M., Jr.; Sahu, R.; Tekwani, B.L.; Duke, S.O.; Khan, S.; Techen, N.; Nanayakkara, N.P.D. Antiplasmodial and cytotoxic cytochalasins from an endophytic fungus, Nemania sp. UM10M, isolated from a diseased Torreya taxifolia leaf. Molecules 2019, 24, 777. [Google Scholar] [CrossRef] [Green Version]
  264. Ateba, J.E.T.; Toghueo, R.M.K.; Awantu, A.F.; Ning, B.M.M.; Gohlke, S.; Sahal, D.; Filho, E.R.; Tsamo, E.; Boyom, F.F.; Sewald, N.; et al. Antiplasmodial Properties and Cytotoxicity of Endophytic Fungi from Symphonia globulifera (Clusiaceae). J. Fungi 2018, 4, 70. [Google Scholar] [CrossRef] [Green Version]
  265. Tawfike, A.F.; Romli, M.; Clements, C.; Abbott, G.; Young, L.; Schumacher, M.; Diederich, M.; Farag, M.; Ebel, R.A.E. Isolation of anticancer and anti-trypanosome secondary metabolites from the endophytic fungus Aspergillus flocculus via bioactivity guided isolation and MS based metabolomics. J. Chromatogr. B 2019, 1106, 71–83. [Google Scholar] [CrossRef] [Green Version]
  266. Santiago, I.M.; Alves, T.M.A.; Rabello, A.; Junior, P.A.S.; Romanha, A.J.; Zani, C.L.; Rosa, C.A.; Rosa, L.H. Leishmanicidal and antitumoral activities of endophytic fungi associated with the Antarctic angiosperms Deschampsia antarctica Desv. and Colobanthus quitensis (Kunth) Bartl. Extremophiles 2012, 16, 95–103. [Google Scholar] [CrossRef]
  267. Fatima, N.; Muhammad, S.A.; Mumtaz, A.; Tariq, H.; Shahzadi, I.; Said, M.S.; Dawood, M. Fungal metabolites and leishmaniasis: A review. Br. J. Pharm. Res. 2016, 12, 101631759. [Google Scholar] [CrossRef]
  268. Fatima, N.; Mumtaz, A.; Shamim, R.; Qadir, M.I.; Muhammad, S.A. In silico analyses of epicoccamides on selected leishmania trypanothione reductase enzyme-based target. Indian J. Pharm. Sci. 2016, 78, 259–266. [Google Scholar] [CrossRef] [Green Version]
  269. Mazlan, N.W.; Tate, R.; Yusoff, Y.M.; Clements, C.; Ebel, R.A.E. Metabolomics-guided isolation of anti-trypanosomal com-pounds from endophytic fungi of the mangrove plant Avicennia lanata. Curr. Med. Chem. 2018, 27, 1815–1835. [Google Scholar] [CrossRef] [PubMed]
  270. Almeida, T.T.D.; Ribeiro, M.A.D.S.; Polonio, J.C.; Garcia, F.P.; Nakamura, C.V.; Meurer, E.C.; Sarragiotto, M.H.; Baldoqui, D.C.; Azevedo, J.L.; Pamphile, J.A. Curvulin and spirostaphylotrichins R and U from extracts produced by two endophytic Bipolaris sp. associated to aquatic macrophytes with antileishmanial activity. Nat. Prod. Res. 2017, 32, 2783–2790. [Google Scholar] [CrossRef] [PubMed]
  271. Ibrahim, S.R.M.; Abdallah, H.M.; Elkhayat, E.S.; Musayeib, N.M.A.; Asfour, H.Z.; Zayed, M.F.; Mohamed, M.A. Fusaripeptide A: New antifungal and anti-malarial cyclodepsipeptide from the endophytic fungus Fusarium sp. J. Asian Nat. Prod. Res. 2017, 20, 75–85. [Google Scholar] [CrossRef] [PubMed]
  272. Ferreira, M.C.; Cantrell, C.L.; Wedge, D.E.; Goncalves, C.N.; Jacob, M.R.; Khan, S.; Rosa, C.A.; Rosa, L.H. Antimycobacterial and antimalarial activities of endophytic fungi associated with the ancient and narrowly endemic neotropical plant Vellozia gigantea from Brazil. Mem. Inst. Oswaldo Cruz. 2017, 112, 692–697. [Google Scholar] [CrossRef] [PubMed]
  273. Metwaly, A.M.; Ghoneim, M.M.; Musa, A. Two new antileishmanial diketopiperazine alkaloids from the endophytic fungus Trichosporum sp. Pharma Chem. 2015, 7, 322–327. [Google Scholar]
  274. Campos, F.F.; Junior, P.A.S.; Romanha, A.J.; Araujo, M.S.S.; Siqueira, E.P.; Resende, J.M.; Alves, T.M.A.; Filho, O.A.M.; Santos, V.L.D.; Rosa, C.A.; et al. Bioactive endophytic fungi isolated from Caesalpinia echinata Lam. (Brazilwood) and identification of beauvericin as a trypanocidal metabolite from Fusarium sp. Mem. Inst. Oswaldo Cruz. 2015, 110, 65–74. [Google Scholar] [CrossRef]
  275. Elkhayat, E.S.; Ibrahim, S.R.M.; Mohamed, G.A.; Ross, S.A. Terrenolide S, a new antileishmanial butenolide from the endophytic fungus Aspergillus terreus. Nat. Prod. Res. 2015, 30, 814–820. [Google Scholar] [CrossRef]
  276. Calcul, L.; Waterman, C.; Ma, W.S.; Lebar, M.D.; Harter, C.; Mutka, T.; Morton, L.; Maignan, P.; Olphen, A.V.; Kyle, D.E.; et al. Screening mangrove endophytic fungi for antimalarial natural products. Mar. Drugs 2013, 11, 5036–5050. [Google Scholar] [CrossRef] [Green Version]
  277. Intaraudom, C.; Boonyuen, N.; Suvannakad, R.; Rachtawee, P.; Pittayakhajonwut, P. Penicolinates A–E from endophytic Penicillium sp. BCC16054. Tetrahedron Lett. 2012, 54, 744–748. [Google Scholar] [CrossRef]
  278. Elfita, E.; Muharni, M.; Munawar, M.; Legasari, L.; Darwati, D. Antimalarial compounds from endophytic fungi of brotowali. (Tinaspora crispa L.). Indones. J. Chem. 2011, 11, 53–58. [Google Scholar] [CrossRef]
  279. Moreno, E.; Varughese, T.; Spadafora, C.; Arnold, A.E.; Coley, P.D.; Kursar, T.A.; Gerwick, W.H.; Rios, L.C. Chemical constituents of the new endophytic fungus Mycosphaerella sp. nov. and their anti-parasitic activity. Nat. Prod. Commun. 2011, 6, 835–840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  280. Li, G.; Lin, P.; Wang, K.; Gu, C.C.; Kusari, S. Artificial intelligence-guided discovery of anticancer lead compounds from plants and associated microorganisms. Trends Cancer 2022, 8, 65–80. [Google Scholar] [CrossRef] [PubMed]
  281. Yang, X.; Wang, Y.; Byrne, R.; Schneider, G.; Yang, S. Concepts of artificial intelligence for computer-assisted drug discovery. Chem. Rev. 2019, 119, 10520–10594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Barre, P.; Stover, B.C.; Muller, K.F.; Steinhage, V. LeafNet: A computer vision system for automatic plant species identification. Ecol. Inform. 2017, 40, 50–56. [Google Scholar] [CrossRef]
  283. Kumar, N.; Belhumeur, P.N.; Biswas, A.; Jacobs, D.W.; Kress, W.H.; Lopez, I.C.; Soares, J.V.B. Leafsnap: A computer vision system for automatic plant species identification. In Computer Vision—ECCV 2012; Springer: Berlin/Heidelberg, Germany, 2012; pp. 502–516. [Google Scholar]
  284. Sun, Y.; Liu, Y.; Wang, G.; Zhang, H. Deep learning for plant identification in natural environment. Comput. Intell. Neurosci. 2017, 2017, 7361042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Clark, C.M.; Costa, M.S.; Sanchez, L.M.; Murphy, B.T. Coupling MALDI-TOF mass spectrometry protein and specialized metabolite analyses to rapidly discriminate bacterial function. Proc. Natl. Acad. Sci. USA 2018, 115, 4981–4986. [Google Scholar] [CrossRef] [Green Version]
  286. Dumolin, C.; Aerts, M.; Verheyde, B.; Schellaert, S.; Vandamme, T.; Jeugt, F.V.D.; Canck, E.D.; Cnockaert, M.; Wieme, A.D.; Cleenwerck, I.; et al. Introducing SPeDE: High-throughput dereplication and accurate determination of microbial diversity from matrix-assisted laser desorption-ionization time of flight mass spectrometry data. mSystems 2019, 4, 1359. [Google Scholar] [CrossRef] [Green Version]
  287. Kautsar, S.A.; Blin, K.; Shaw, S.; Navarro-Munoz, J.C.; Terlouw, B.R.; Hooft, J.J.J.V.D.; Santen, J.A.V.; Tracanna, V.; Duran, H.G.S.; Andreu, V.P.; et al. MIBiG 2.0: A repository for biosynthetic gene clusters of known function. Nucleic Acids Res. 2019, 48, 454–458. [Google Scholar] [CrossRef] [Green Version]
  288. Palaniappan, K.; Chen, I.M.A.; Chu, K.; Ratner, A.; Seshadri, R.; Kyrpides, N.C.; Ivanova, N.N.; Mouncey, N.J. IMG-ABC v.5.0: An update to the IMG/atlas of biosynthetic gene clusters knowledgebase. Nucleic Acids Res. 2019, 48, 422–430. [Google Scholar] [CrossRef]
  289. Blin, K.; Shaw, S.; Steinke, K.; Villebro, R.; Ziemert, N.; Lee, S.Y.; Medema, M.H.; Weber, T. antiSMASH 5.0: Updates to the secondary metabolite genome mining pipeline. Nucleic Acids Res. 2019, 47, 81–87. [Google Scholar] [CrossRef] [Green Version]
  290. Ricart, E.; Pupin, M.; Muller, M.; Lisacek, F. Automatic annotation and dereplication of tandem mass spectra of peptidic natural products. Anal. Chem. 2020, 92, 15862–15871. [Google Scholar] [CrossRef] [PubMed]
  291. Kurita, K.L.; Glassey, E.; Linington, R.G. Integration of high-content screening and untargeted metabolomics for comprehensive functional annotation of natural product libraries. Proc. Natl. Acad. Sci. USA 2015, 112, 11999–12004. [Google Scholar] [CrossRef] [Green Version]
  292. Grienke, U.; Foster, P.A.; Zwirchmayr, J.; Tahir, A.; Rollinger, J.M.; Mikros, E. 1H NMR-MS-based heterocovariance as a drug discovery tool for fishing bioactive compounds out of a complex mixture of structural analogues. Sci. Rep. 2019, 9, 11113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Zani, C.L.; Carroll, A.R. Database for rapid dereplication of known natural products using data from MS and fast NMR experiments. J. Nat. Prod. 2017, 80, 1758–1766. [Google Scholar] [CrossRef] [PubMed]
  294. Phosrithong, N.; Ungwitayatorn, J. Molecular docking study on anticancer activity of plant-derived natural products. Med. Chem. Res. 2010, 19, 817–835. [Google Scholar] [CrossRef]
  295. Sharma, V.; Sharma, P.C.; Kumar, V. In silico molecular docking analysis of natural pyridoacridines as anticancer agents. Adv. Chem. 2016, 2016, 409387. [Google Scholar] [CrossRef] [Green Version]
  296. Reker, D.; Rodrigues, T.; Schneider, P.; Schneider, G. Identifying the macromolecular targets of de novo-designed chemical entities through self-organizing map consensus. Proc. Natl. Acad. Sci. USA 2014, 111, 4067–4072. [Google Scholar] [CrossRef] [Green Version]
  297. Madhukar, N.S.; Khade, P.K.; Huang, L.; Gayvert, K.; Galletti, G.; Stogniew, M.; Allen, J.E.; Giannakakou, P.; Elemento, O. A Bayesian machine learning approach for drug target identification using diverse data types. Nat. Commun. 2019, 10, 5221. [Google Scholar] [CrossRef] [Green Version]
Figure 1. A schematic representation of plant–fungus cost–benefit interactions including interactions among host plant, endophytic fungi, pathogens, and insects which leads fungal endophytes to produce an array of metabolites with profound bioactivities.
Figure 1. A schematic representation of plant–fungus cost–benefit interactions including interactions among host plant, endophytic fungi, pathogens, and insects which leads fungal endophytes to produce an array of metabolites with profound bioactivities.
Biomolecules 13 01038 g001
Figure 2. Structures of plant-analogous compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 2. Structures of plant-analogous compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g002
Figure 3. Structures of anticancer compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 3. Structures of anticancer compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g003
Figure 4. Structures of antioxidant compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 4. Structures of antioxidant compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g004
Figure 5. Structures of anti-inflammatory compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 5. Structures of anti-inflammatory compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g005
Figure 6. Structures of antidiabetic compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 6. Structures of antidiabetic compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g006
Figure 7. Structures of immunosuppressive compounds from fungal endophytes (structues were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 7. Structures of immunosuppressive compounds from fungal endophytes (structues were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g007
Figure 8. Structures of antibacterial compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 8. Structures of antibacterial compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g008
Figure 9. Structures of antifungal compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 9. Structures of antifungal compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g009
Figure 10. Structures of antiviral compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 10. Structures of antiviral compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g010
Figure 11. Structures of antiprotozoal compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Figure 11. Structures of antiprotozoal compounds from fungal endophytes (structures were taken from protologue publications and were redrawn using ChemDraw Ultra 12.0).
Biomolecules 13 01038 g011
Table 1. Fungal endophytes producing bioactive compounds (and their derivatives) analogous to their host plants.
Table 1. Fungal endophytes producing bioactive compounds (and their derivatives) analogous to their host plants.
CompoundFungal EndophyteHost PlantBioactivityReferences
TaxolTaxomyces andreanaeTaxus brevifoliaCytotoxic[62]
Pestalotiopsis microsporaTaxus wallachianaCytotoxic[76]
Tubercularia sp. TF5Taxus maireiCytotoxic[77]
Fusarium redolensTaxus baccataAntimitotic[78]
VinblastineAlternaria sp.Catharanthus roseus-[66]
Fusarium oxysporumCatharanthus roseus-[79]
Nigrospora sphaericaCatharanthus roseusCytotoxic[80]
VincristineFusarium oxysporumCatharanthus roseus-[81]
Fusarium oxysporum AA-CRL-6Catharanthus roseus-[79]
Talaromyces radicus CrP20Catharanthus roseusCytotoxic[82]
Eutypella sp. CrP14Catharanthus roseusCytotoxic[83]
CamptothecinEntrophospora infrequensNothapodytes foetidaCytotoxic[67]
Neurospora crassaNothapodytes foetidaCytotoxic[84]
Nodulisporium sp.Nothapodytes foetida-[85]
Fusarium solaniCamptotheca acuminata-[61]
Trichoderma atroviride LY357Camptotheca acuminata-[86]
Fusarium solani S-019Camptotheca acuminataCytotoxic[87]
PodophyllotoxinPhialocephala fortinii (PPE5 and PPE7)Podophyllum peltatumCytotoxicity[68]
Trametes hirsuteSinopodophyllum hexandrumCytotoxic[88]
Mucor fragilis TW5Sinopodophyllum hexandrum-[89]
HuperzineShiraia sp. Slf14Huperzia serrataAcetylcholinesterase inhibition[90]
Cladosporium cladosporioides LF70Huperzia serrataAcetylcholinesterase inhibition[91]
Paecilomyces tenuis YS-13Huperzia serrataAcetylcholinesterase inhibition[92]
Trichoderma sp. L44Huperzia serrataAcetylcholinesterase inhibition[93]
HypericinThielavia subthermophilaHypericum perforatumAntimicrobial, cytotoxic[69]
ResveratrolAlternaria sp. MG1Vitis vinifera-[94]
Quambalaria cyanescensVitis viniferaAntibacterial, antioxidant, cytotoxic[20]
Table 2. Anticancer compounds produced by fungal endophytes.
Table 2. Anticancer compounds produced by fungal endophytes.
Fungal EndophyteHost PlantBioactive CompoundsTested Cell LinesIC50 or Inhibition (%)References
Alternaria alternataJatropha heyneiKaempferolLung carcinoma cancer cell line (A549)393.52 µg/mL[108]
Aspergillus flavusCynodon dactylon2,4,7-trinitrofluorenone and 22t-triene-6beta-olMCF-7 breast cancer cell line16.25 μg/mL[109]
Quambalaria cyanescensVitis viniferaResveratrolA549 cell line82%[20]
Penicillium citrinum CGJ-C2Tragia involucrataQuercetinMCF-7 cell line1 µg/mL[110]
J-1, J-2, and J-3Ginkgo bilobaPodophyllotoxinHeLa cell lines75%[111]
Simplicillium subtropicum SPC3Duguetia staudtiiSimplicilones A and BCervix carcinoma cell line KB3.125–29 μg/mL[112]
Trichoderma virideZiziphus mauritiana3-beta-hydroxyurs-12-en-28-oic acidHeLa cell lines23 μg/mL[113]
Xylaria sp. ZJWCF255Ficus caricaCytochalasin QSMMC-772, MCF-7, MGc 80-3 cell lines7–17 μg/mL[34]
Phomopsis sp. BCC 45011Xylocarpus granatumPhomoxydiene C and Cytosporone EKB, MCF-7, NCI-H187, Vero cells1.49–40.17 μg/mL[34]
Pestalotiopsis uvicolaArtemisia japonicaKaempferol, Quercetin, Rutin, GenisteinAdriamycin-resistant (ADR) MCF-7, ADR, and
ovarian paclitaxel-resistant cell A2780 cells
-[34]
Pestalotiopsis sp. FT172Myrsine sandwicensis(+)-ambuic acidCisplatin-resistant A2780 cell lines3–17 μM[34]
A. alternata MGTMMP031Vitex negundoAlternariol methyl etherHepatocellular carcinoma HepG2-[114]
Aspergillus terreusBruguiera gymnorrhyzaCowabenzophenone AColon cancer cell line10 μM[115]
Chaetomium globosumCouroupita guianensisFlavipinA549, colorectal adenocarcinoma cells (HT-29), MCF-7 cancer cell lines9–54 µg/mL[116]
Fusarium solaniCamptotheca acuminataCamptothecinVero, prostatic adenocarcinoma cells (PC-3) cells-[87]
Sir-SM2Annona muricataHexahydro-3-(2-methylpropyl)-Pyrrolo[1,2-a]pyrazine-1,4-dioneWiDr cell lines20 µg/mL[117]
Aspergillus terreusAchyranthus asperaTerrein (4,5-Dihydroxy-3-(1-propenyl)-2-cyclopenten-1-one)A-549121 µg/mL[118]
Xylaria psidiiAegle marmelos(−) 5-methylmelleinMCF-7, MIA-Pa-Ca-2, NCI-H226, HepG2, and DU14516–37 μM[104]
Sordariomycetes sp. (PDA)BL5Strobilanthes crispusPyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(2-methylpropyl)PC-3, HepG2, A-549, HT-29, MCF-727–161 µg/mL[119]
Pestalotiopsis sp.Dendrobium officinale(4S,6S)-6-[(1S,2R)-1,2-dihydroxybutyl]- 4-hydroxy-4-methoxytetrahydro-2H-pyran-2-one, (6S,2E)-6-hydroxy-3-methoxy-5-oxodec-2-enoic acidHL-60 cell lines183 μM[120]
Pseudolagarobasidium acaciicolaBruguiera gymnorrhizMerulin B and C, steperoxide AHL-60, HepG20.08–49.08 µg/mL[121]
A. terreusCodium decorticatumF8HepG27 µg/mL[122]
C. globosumGinkgo bilobaChaetoglobosin AColon cancer cell lines (HCT116)3–8 µM[107]
Myrothecium roridumAjuga decumbensMyrotheciumones AHepG25 µM[123]
Phomopsis glabraePongamia pinnataDepsipeptide (PM181110)BXFT24, CXF 269L (colon), LXFA 629L (lung), PAXF 546L cell lines0.04–0.055 µM[124]
Hypocrea lixiiCajanus cajanCajanolA54920 µg/mL[125]
Penicillium janthinellum Yuan-27Panax ginsengBrefeldin AMKN45, LOVO, A549, MDA-MB-435, HepG2, HL-60 cell lines0.49–7.46 μg/mL[126]
Fomitopsis sp. (MTCC 10177) Miquelia dentataCamptothecinHCT-116, SW-4805–23 μg/mL[127]
Fusarium proliferatum (MTCC 9690)Dysoxylum binectariferumRohitukineHCT-116, MCF-7 cancer cell lines10 μg/mL[128]
Table 3. Antioxidant compounds produced by fungal endophytes.
Table 3. Antioxidant compounds produced by fungal endophytes.
Fungal EndophyteHost PlantBioactive CompoundsIC50 or Inhibition (%)References
PenicilliumcitrinumDigitaria bicornisDL-carnitine, α-Eleostearic acid, Benzophenone, Sclerotiorin, Cafeic acid, Oleamide, Stearamide0.76–55 µg/mL[38]
Penicillium decumbens-Sulforhodamine B-[146]
Aspergillus tubenginses ASH4Hyoscyamus muticusAnofinic acid-[134]
ZA 163, MO 211, LO 261, FE 082, and FE 084Albizia zygia,
Millettia thonningii, Alchornea cordifolia, Ficus exasperat
Pyrogallol, Di-alpha-tocopherol, Alpha tocospiro, Linoleic acid, 9-octadecenamide, Lupeol, and 9-octadecenoic acid (Z)-[14]
Aspergillus nidulans, Aspergillus fumigatus, Aspergillus favusOcimum basilicum9-Octadecenoic acid (Z)-, Hexadecanoic acid, 1-nonadecene68–347 µg/mL[147]
A. fumigatusMoringa oleiferaCafeic acid, Rutin, Ellagic acid, Quercetin, Kaempferol40 µg/mL[36]
Chaetomium globosum, Aspergillus nidulansPassifora incarnataMethoxymethylphenol, Orcinol, Sorbicillin0.21–0.324 mg/mL[148]
C. globosumMoringa oleiferaCatechin, Chlorogenic acid, Cafeic acid, Umbelliferone, Coumaric acid, Kaempferol45–50 µg/mL[36]
C. globosumConyza blinii3-methoxyflavone, Nobiletin, Scopoletin, and Daidzein0.01–0.11 mg/mL[149]
Alternaria sp. Samif01Salvia miltiorrhiza3-epi-dihydroaltenuene A, Altenuisol, 4-hydroxyalternariol-9-methyl ether474 μM[150]
Nigrospora sp.O. basilicumNezukol, Z-lanceol, Chavicol, Catalponone15 μg/mL[135]
Talaromyces islandicus EN-501Laurencia okamurai8-hydroxyconiothyrinone B, 8,11-dihydroxyconiothyrinone B61 μM[35]
Pseudocercospora sp. ESL 02Elaeocarpus sylvestrisTerreic acid and 6-methylsalicylic acid30 mg/mL[151]
Epicoccum nigrumEntada abyssinicaQuinizarin11 μg/mL[152]
A. fumigatesCajanus cajanLuteolin22 μg/mL[153]
Table 4. Anti-inflammatory compounds produced by fungal endophytes.
Table 4. Anti-inflammatory compounds produced by fungal endophytes.
Fungal EndophyteHost PlantBioactive CompoundsInhibitionIC50 or Inhibition (%)References
Aspergillus nigerElaeocarpus floribundusAsnipyrone B, Hexylitaconic acid, Chlorogenic acid, Nigragillin, FusarubinNO, COX-II, IL-1, IL-6, and TNF-α-[40]
Diaporthe sp. QYM12Kandelia candelDiaporpenoid A, Diaporpyrones ANO12–21 µM[161]
Talaromyces sp. SK-S009Kandelia obovata6-[1-(acetyloxy) ethyl)] 5-hydroxy-2,7-dimethoxy-1,4-napthalenedioneNO1.7 µM[162]
Edenia gomezpompaeUnidentified plantPreussomerin EG1NO-[163]
Phomopsis sp. SYSUQYP-23Kandelia candelFarinomalein A, B, and H, PhenylahistinNO15–25 μM[164]
Fusarium chlamydosporumAnvillea garciniiChlamydosterol A and B5-lipoxygenase3.57 μM[165]
Aspergillus sp.Trichocoleaceae sp.6-hydroxy-3-methoxyviridicatin, notoamide B, 3-O-methylviridicatolNO22–50 μM[166]
Fusarium sp.Mentha longifoliaFusaristerols B5-lipoxygenase2–4 μM[167]
A. terreusStrigamia maritimaAsperimides C and DNO1.26 μM[168]
Ascomycota sp. CYSK-4Pluchea indicaDesmethyldichlorodiaportintoneNO15.8 µM[169]
Lasiodiplodia theobromae ZJ-HQ1Acanthus ilicifoliusLasiodiplactone ANO23.5 μM[170]
Trichoderma sp. Xy24Xylocarpus granatumCyclonerodiol BNO75%[155]
A. terreus PR-P-2Camellia sinensisAsperteretal C, butyrolactone INO16–27 μM[171]
Botryosphaeria sp. SCSIO KcF6Kandelia candelBotryosphaerin-BCOX-21.12 μM[155]
Periconia sp.Annonsa muricataPericonianone ANO0.15–0.38 μM[172]
Dendryphion nanumFicus religiosaHerbarinCytokines TNF-α and IL-60.60 µM[158]
NO: nitric oxide, COX: cyclooxygenase, TNF: tumor necrosis factor.
Table 5. Anti-diabetic compounds produced by fungal endophytes.
Table 5. Anti-diabetic compounds produced by fungal endophytes.
Fungal EndophyteHost PlantBioactive CompoundsInhibitionIC50 or Inhibition (%)References
Aspergillus sp.Sonneratia apetalaAspergiamides A, Fα-glucosidase40–83 µM[173]
Penicillium canescensJuniperus polycarposMethylxanthoneα-glucosidase32 μM[184]
Xylariaceae sp. QGS01Querus gilva8-Hydroxy-6,7-dimethoxy-3-methylisocoumarineα-glucosidase41.75 μg/mL[42]
Nigrospora oryzaeCombretum dolichopetalumS (+)-2 cis-4-trans-abscisic acid, 7-hydroxy-abscisic acid, 4-des-hydroxy altersolanol ADiabetic-
induced mice
30–46%[173]
Epicoccum sp. HS-1Apostichopus japonicusIsopimarane diterpeneα-glucosidase4.6–11.9 μM[183]
Aspergillus awamoriAcacia niloticaPeptidesα-amylase, α-glucosidase3–6 μg/mL[176]
Chaetomiaceae sp. MEXU 27095Hintonia latifloraThielavins A, J, and Kα-glucosidase15–23 μM[174]
Xylaria sp.-Eremophilane sesquiterpenesα-glucosidase6.54 μM[181]
Table 6. Immunosuppressive compounds produced by fungal endophytes.
Table 6. Immunosuppressive compounds produced by fungal endophytes.
Fungal EndophyteHost PlantBioactive CompoundsInhibitionPercentage Inhibition or IC50References
Albifmbria viridisCoptis chinensisAlbifpyrrol BLPS (B cells)16.16 μM[47]
Ilyonectria robustaBletilla striataRobustaditerpene C and EConcanavalin (Con) A (T cells) and LPS (B cells)17–75 μM[194]
Aspergillus sp.Tripterygium wilfordiiPseurotinanti-CD3/anti-CD28 mAbs8–9 μM[46]
Daldinia sp. TJ403-LS1Anoectochilus roxburghiiDaldiniol ALPS and antiCD3/anti-CD28 mAbs0.06 μM[195]
Aspergillus fumigatusCynodon dactylonBisdethiobis (methylthio) Gliotoxin, Fumitremorgin C, 3-(hydroxyacetyl) indoleCon A (T cells) and LPS (B cells)1.08–97 μM[45]
Fusarium sp. and Cladosporium sp.Psidium guajava and Newbouldia laevisCitrinin, Nidulalin, p-hydroxybenzoic acid, Cyclopenin--[188]
Pestalotiopsis sp. HHL-14Rhizophora stylosaPhomoxydiene C, Z-isomer, mycoepoxydieneCon A (T cells) and LPS (B cells)33–97 μM[196]
A. fumigatus HQD24Rhizophora mucronata16-O-deacetylhelvolic acid 21,16-lactoneCon A (T cells) and LPS (B cells)12–62 μM[197]
Mycospaerella nawae ZJLQ129Smilax china(−) mycousnineAntigens CD25 and CD69-[189]
Penicillium sp. ZJ-SY2Sonneratia apetalaPeniphenone, Conioxanthone A, Pinselin, Sydowinin ACon A (T cells) and LPS (B cells)6–9 µg/mL[190]
Fusarium subglutinansTripterygium wilfordiiSubglutinol A and BProinflammatory IFNγ and IL-17-[191]
Phomopsis sp. S12Illigera rhodanthaLibertellenone JLPS3–15 μM[192]
Aspergillus sydowiiScapania ciliataEmodinCon A- and LPS8–10 µg/ml[44]
LPS: lipopolysaccharides, IFN: interferon, IL: interleukin.
Table 7. Antimicrobial compounds produced by fungal endophytes.
Table 7. Antimicrobial compounds produced by fungal endophytes.
Fungal EndophyteHost PlantBioactive CompoundsTested Pathogen(s)IC50 or Inhibition (%)References
Antibacterial compounds
Aspergillus fumigatusCeriops decandraFumigaclavine C, Azaspirofuran B, FraxetinStaphylococcus aureus, Micrococcus luteus, Escherichia coli, Pseudomonas aeruginosa0.078–5 mg/mL[204]
Emericella sp.Panax notoginsengQuiannulatic acidMultidrug-resistant (MDR) Enterococcus faecium12.5 µg/mL[206]
Aspergillus nigerOpuntia ficus-indicaCristatumin B, DihydroauroglaucinMDR S. aureus, E. faecalis, Klebsiella. pneumonia, P. aeruginosa2–125 µg/mL[205]
Penicillium citrinumDigitaria bicornisCiprofloxacinS. aureus, Salmonella typhi, E. faecalis, E. coli9–20%[203]
Curvularia papendorfiiVernonia amygdalinaPolyhydroxyacid, Kheiric acidMethicillin resistant S. aureus (MRSA)62.5 µg/mL[208]
Aspergillus cejpiiNelumbo nucifera5-(1H-Indol-3-yl)-4,5-dihydro-[1,2,4]triazin-3-ylamineMRSA-[209]
Diaporthe sp.Pteroceltis tatarinowiiDiaporone ABacillus subtilis66.7 μM[202]
Curvularia lunataPaepalanthus chiquitensisCurvulinic acidE. coli62.5 μg/mL[210]
Alternaria alternata AE1Azadirachta indicaPhenanthrene, 7-isopropyl-1-methyl (Retene), DichloronitromethaneB. subtilis, Listeria monocytogenes, S. aureus, E. coli, Salmonella typhimurium-[136]
Fusarium sambucinum TE-6LNicotiana tabacumAmoenamide C, Sclerotiamide BE. coli, M. luteus, P. aeruginosa4–8 μg/mL[211]
Curvularia sp. T12Rauwolfia macrophylla2′-deoxyribolactone, Hexylitaconic acid, ErgosterolPseudomonas agarici, E. coli, Staphylococcus warneri, M. luteus-[212]
Arthrinium sp. MFLUCC16-1053Zingiber cassumunar3-p-menthone, Bornyl acetate, γ-curcumene, Bicyclogermacrene, and β-isocomeneS. aureus, E. coli7–31 µg/mL[213]
Aspergillus clavatonanicus MJ31Mirabilis jalapa6-PP (6 Pentyl-2H Pyrone-2-one), 1, 2 butadiene, m-camphorene, 3-Thietanol, Thiopivalic acid, Pthalic acid, Heneicosane, Pyrazol, and benzene derivativesE. coli, S. aureus, M. luteus, B. subtilis0.078–0.62 mg/mL[201]
Colletotrichum sp. BS4Buxus sinicaColletotrichone A, B, C and Chermesinone BS. aureus, E. coli, B. subtilis, P. aeruginosa>10 µg/mL[214]
Lasiodiplodia theobromae ZJ-HQ1Acanthus ilicifoliusChloropreussomerin A and B, Preussomerin MS. aureus, B. subtilis, E. coli, P. aeruginosa, Salmonella enteritidis1–13 μg/mL[215]
Epicoccum nigrumEntada abyssinicaQuinizarin, indole-3-carboxylic acid, and ParahydroxybenzaldehydeB. cereus, S. typhimurium3–6.25 μg/mL[152]
Emericella qaudrilineata (RS-5)Pteris pellucidBenzyl benzoate, Benzaldehyde dimethyl acetal, and Benzoic acidS. aureus, Aeromonas hydrophilla-[216]
Penicillium brocae MA-231Avicennia marinaPenicibrocazine A–ES. aureus, M. luteus, Gaeumannomyces graminis0.25–64 μg/mL[217]
Fusarium sp.Opuntia dilleniiEquisetinB. subtilis, MRSA8–16 μg/mL[218]
Microsphaeropsis sp., Seimatosporium sp.Salsola oppositifoliaMicrosphaerol, SeimatoroneE. coli, Bacillus megaterium8–9 mg/mL[219]
Colletotrichum sp.UmbelliferaeColletonoic acidB. megaterium8 mg/mL[220]
Eupenicillium sp. LG41Xanthium sibiricumEupenicinicols A and B, (2S)-butylitaconic acid,
(2S)-hexylitaconic acid
B. subtilis, S. aureus, E. coli, Acinetobacter sp.1- > 10.0 μg/mL[221]
Botryosphaeria dothidea X-4Camptotheca acuminata9-methoxycamptothecinB. subtilis, E. coli,47.6%[222]
Phoma sp. PG23Taraxacum mongolicum2-hydroxy-6-methylbenzoic acidE. coli, S. aureus, A. hydrophila, Edwardsiella tarda, Pasteurella multocida-[223]
Gliomastix murorum Ppf8Paris polyphyllaErgosta-5,7,22-trien-3-ol, 2,3-dihydro-5-hydroxy-α,α-dimethyl-2-benzofuranmethanolE. coli, Pseudomonas lachrymans, B. subtilis, Staphylococcus haemolyticus65–146 µg/mL[224]
Penicillium chrysogenumPorteresia coarctataDipodazine DVibrio cholerae-[225]
Diaporthe phaseolorumLaguncularia racemosa3-hydroxypropionic acidS. aureus, S. typhi64 µg/mL[226]
Aspergillus sp. EJC08Bauhinia guianensisFunigaclavine C, Pseurotin AB. subtilis, E. coli, P. aeruginosa, S. aureus7–31 µg/mL[227]
Hyalodendriella sp. Ponipodef12Populus deltoidesPalmariol B, 4-hydroxymellein, Alternariol 9-methyl etherB. subtilis, P. lachrymans16–19 µg/mL[228]
Phomopsis longicollaBostrychia radicansDicerandrol CS. aureus1.33 µg/mL[229]
Pestalotiopsis mangiferaeMangifera indica4-(2,4,7-trioxa-bicyclo[4.1.0]heptan-3-yl)B. subtilis, K. pneumoniae, E. coli, M. luteus, P. aeruginosa-[230]
Pestalotiopsis sp.Arbutus unedoPestalotheol G, Anofinic acidE. coli, B. megaterium7–12 µg/mL[231]
Phomopsis sp.Cistus monspeliensisPhomochromone A-B, Phomotenone, (1S,2S,4S)-trihydroxy-p-menthaneE. coli, B. megaterium6–8 µg/mL[232]
Phomopsis sp.Allamanda catharticaTerpeneS. aureus, B. subtilis, S. typhi-[233]
Antifungal compounds
Curvularia protuberatePaspalidium favidumDiphenyl sulfone, 7-Hydroxycoumarine, Griseofulvin, β-AsaroneAlternaria alternata, Fusarium oxysporum31–62 µg/mL[203]
Cladosporium cladosporioidesZygophyllum mandavillei3-phenylpropionic acidAspergillus flavus and Fusaroum solani3.90–15.62 mg/mL[234]
Aplosporella javeedii, Orychophragmus violaceusAplojaveediins A–FCandida albicans ATCC 24433-[235]
Alternaria tenuissima OE7Ocimum tenuiflorum1,2-PentanediolC. albican100–500 µg/mL[136]
P. citrinumStephania kwangsiensisCitrinin, EmodinAlternaria citri3 μg/mL[236]
F. oxysporum KU527806Dendrobim lindleyGibepy- rone A, Pyrrolo[1,2-a] pyrazine-1, 4-dione, hexahydro-3-(2-methylpropyl), and in- doleacetic acidC. albicans, C. tropicalis, Curvularia sp., f. sp.-[237]
Cladosporium delicatulumTerminalia pallida, Rhychosia beddomei, Pterocarpu santalinusPlumbagin (5-hydroxyl- 2- methylnaptalene-1,4-dione)C. albicans, C. tropicalis, F. moniliforme6–12 mg/mL[238]
Aspergillus flavusLannea coromandelicaKojic acid, Octadecanoic acid, Diethyl phylate, 3-Phenyl propionic acidC. ablicans, Malassezia pachydermis-[239]
Mycosphaerella sp. UFMGCB 2032Eugenia bimarginata(2S,3R,4R)-(E)-2-amino-3,4-Antifungal dihydroxy-2-(hydroxymethyl)-14-oxoeicos6,12-dienoic acid, and MyriocinCryptococcus neoformans, Cryptococcus gattii7–31 µg/mL[240]
Lophodermium nitens DAOM 250027Pinus strobus(7R)-(-)-methoxysydonol and its derivatives, (7R,7′R)-(-)-pyrenophorinSaccharomyces cerevisiae5 µM[199]
Phialophora musteaCrocus sativusPhialomustinC. albicans73.6 µM[241]
Trichothecium sp.Phyllanthus amarusTrichothecinol-ACryptococcus albidus25 µg/mL[242]
Lophodermium sp.Pinus strobusMethyl (2Z,4E)-6(acetyloxy)-5-formyl-7-oxoocta-2,4-dienoate, 5-(hydroxymethyl)-2-(20, 60, 60-trimethyltetrahydro-2H-pyran-2- yl)phenol, pyrenophorolS. cerevisae2 μM[243]
Massrison sp.Rehmannia glutinosaMassarigenin D, Spiromassaritone, PaecilospironeC. albicans, C. neoformans, T. rubrum,
A. fumigatus
1–4 μg/mL[244]
Antiviral compounds
A. terreusGlycine maxAspulvinone EHIV-[245]
Phoma sp.YE3135Aconitum vilmorinianumPhomanolideH1N12–20 μg/mL[246]
Pleospora tardaEphedra aphyllaAlternariol and Alternariol-(9)-methyl etherHSV15–40%[145]
Hypoxylon sp. 6269Artemisia annuaIntegracide E and Isointegracide EHIV31–100 μM[247]
Aspergillus sp. CPCC 400735Kadsura longipedunculataAsperphenalenone A and DHIV2–9 μM[248]
Nigrospora sp. YE3033Aconitum carmichaeli6-O-demethyl-4-dehydroxyaltersolanol A, 4-dehydroxyaltersolanol A, Altersolanol B, Cermesinone BH1N10.80–8 µg/mL[249]
Pestalotiopsis theaFagara zanthoxyloidesChloroisosulochrin ficipyrone A, and Pestheic acidRSV0.57–2 µg/mL[250]
Periconia sp. F-31Annona muricataPericoannosin A, Periconiasins FHIV67 μM[75]
Alternaria sp.Calophyllum inophyllumCoumarinHIV-[251]
Chaetomium globosum TW1-1Armadillidium vulgareArmochaetoglobins K–RHIV0 0.11–0.55 μM[252]
Cercosporella sp.Schisandra chinensisErgosterol Peroxide, Ergosterol, β-Sitosterol, StigmasterolHIV-[253]
A. tenuissima QUE1SeQuercus emoryiAltertoxins V, Altertoxins I-IIIHIV0.5–2 μg/mL[254]
A. tenuissimaQuercus emoryiDK, DL, DM, and DPHIV1.5 μg/mL[255]
Emericella sp. (HK-ZJ)Aegiceras corniculatumEmerimidine A–B and Emeriphenolicins A–DH1N150%[256]
Table 8. Anti-protozoan compounds produced by fungal endophytes.
Table 8. Anti-protozoan compounds produced by fungal endophytes.
Fungal EndophyteHost PlantBioactive CompoundsInhibitionIC50 or Inhibition (%)References
Nigrospora oryzae CF-298113Triticum sp.PipecolisporinPlasmodium falciparum, Trypanosoma cruzi3.21 µM[260]
Nemania sp. UM10MTorreya taxifolia19,20-epoxycytochalasins CP. falciparum, P. berghei0.05 µM[263]
Fusarium sp., Lasiodiplodia theobromaeAvicennia lanataAnhydrofusarubin, Javanicin, Dihydrojavanicin, Solaniol, (-)-melleinTrypanosoma brucei brucei0.047–0.276 µg/mL[269]
Aspergillus flocculusMarkhamia platycalyxErgosterol, Ergosterol peroxideT. brucei brucei7.3–31.6 μM[265]
Bipolaris sp. C36, Bipolaris sp. AZ26Deschampsia antarctica, Colobanthus quitensisCurvulin, Spirostaphylotrichin R, ULeishmania amazonensis70–84 µg/mL[270]
Fusarium sp.Mentha longifoliaFusaripeptide AP. falciparum0.34 μM[271]
Diaporthe miriciaeVellozia giganteaEpoxicitocalasin HP. falciparum chloroquine-sensitive and resistant strains39–51 µg/mL[272]
E. nigrum-EpicoccamideLeishmania sp.-[268]
Trichosporum sp.Trigonella foenum-graecum(6-S)-3-(1,3-dihydroxypropyl)-6-(2-methylpropyl)piperazine-2,5-dione
(6-R)-3-(1,3-dihydroxypropyl)-6-(2methylpropyl)piperazine-2,5-dione
Leishmania donovani82–96 µg/mL[273]
Fusarium sp. WC 9Caesalpinia echinataBeauvericinT. cruzi1.9 μg/mL[274]
A. terreusCarthamus lanatus(22E,24R)-stigmasta-5,7,22-trien-3-β-ol, stigmast-4-ene-3-one, terrenolide SL. donovani15–27 µM[275]
Diaporthe sp. CY-5188Kandelia obovate, Avicennia marinaDicerandrol DP. falciparum-[276]
Penicillium sp. BCC16054GrassPenicolinates A–CP. falciparum3.07–3.25 µg/mL[277]
BB1-BB5, BD4-BD6Tinaspora crispa2,5-dihydroxy-1-(hydroxymethyl)pyridin4-onP. falciparum0.129 µM[278]
Mycosphaerella sp. F2140,Psychotria horizontalisCercosporinP. falciparum, L. donovani,
T. cruzi
0.46–1.08 µM[279]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gupta, A.; Meshram, V.; Gupta, M.; Goyal, S.; Qureshi, K.A.; Jaremko, M.; Shukla, K.K. Fungal Endophytes: Microfactories of Novel Bioactive Compounds with Therapeutic Interventions; A Comprehensive Review on the Biotechnological Developments in the Field of Fungal Endophytic Biology over the Last Decade. Biomolecules 2023, 13, 1038. https://doi.org/10.3390/biom13071038

AMA Style

Gupta A, Meshram V, Gupta M, Goyal S, Qureshi KA, Jaremko M, Shukla KK. Fungal Endophytes: Microfactories of Novel Bioactive Compounds with Therapeutic Interventions; A Comprehensive Review on the Biotechnological Developments in the Field of Fungal Endophytic Biology over the Last Decade. Biomolecules. 2023; 13(7):1038. https://doi.org/10.3390/biom13071038

Chicago/Turabian Style

Gupta, Aditi, Vineet Meshram, Mahiti Gupta, Soniya Goyal, Kamal Ahmad Qureshi, Mariusz Jaremko, and Kamlesh Kumar Shukla. 2023. "Fungal Endophytes: Microfactories of Novel Bioactive Compounds with Therapeutic Interventions; A Comprehensive Review on the Biotechnological Developments in the Field of Fungal Endophytic Biology over the Last Decade" Biomolecules 13, no. 7: 1038. https://doi.org/10.3390/biom13071038

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

Article Metrics

Back to TopTop