Next Article in Journal
Methylation-GC-MS/FID-Based Glycosidic Linkage Analysis of Unfractionated Polysaccharides in Red Seaweeds
Previous Article in Journal
Inhibition Effects and Mechanisms of Marine Compound Mycophenolic Acid Methyl Ester against Influenza A Virus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Bioactive Natural Products from Marine-Derived Penicillium Fungi: A Review (2021–2023)

by
Fang Lv
1 and
Yanbo Zeng
2,*
1
Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
2
Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources & National Key Laboratory for Tropical Crop Breeding, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
*
Author to whom correspondence should be addressed.
Mar. Drugs 2024, 22(5), 191; https://doi.org/10.3390/md22050191
Submission received: 12 March 2024 / Revised: 13 April 2024 / Accepted: 22 April 2024 / Published: 23 April 2024

Abstract

:
Marine-derived Penicillium fungi are productive sources of structurally unique and diverse bioactive secondary metabolites, representing a hot topic in natural product research. This review describes structural diversity, bioactivities and statistical research of 452 new natural products from marine-derived Penicillium fungi covering 2021 to 2023. Sediments are the main sources of marine-derived Penicillium fungi for producing nearly 56% new natural products. Polyketides, alkaloids, and terpenoids displayed diverse biological activities and are the major contributors to antibacterial activity, cytotoxicity, anti-inflammatory and enzyme inhibitory capacities. Polyketides had higher proportions of new bioactive compounds in new compounds than other chemical classes. The characteristics of studies in recent years are presented.

1. Introduction

Marine-derived fungi have a variety of medical applications due to their capability of generating various enzymes and antimicrobial agents [1]. Since the first species Sphaeria posidoniae (Halotthia posidoniae) on the rhizome of the sea grass Posidonia oceanica was studied in 1846 [2], scientists have never stopped studying the natural products (NPs) of marine-derived fungi [3]. The rapid development of marine bio-technology and ever-increasing needs of clinic applications resulted in the emergence of marine natural products as alternative drug sources in the early 1990s [4] and the voluminous output in natural product research from the fungi isolated from different marine animals, seaweeds and sediments, with many new bioactive compounds being described each year [5].
As an essential part of marine micro-organisms, Penicillium fungi have received great attention among all marine-derived fungi, accounting for 22% of NPs of marine fungal origin, and play an important role in the discovery of marine natural products with bioactivities and novel structures [6]. Several reviews on natural products isolated from marine-derived Penicillium species have been published [4,6,7,8]. A total of 390 new secondary metabolites from marine-derived Penicillium fungi were highlighted from 1991 to 2014 [6], and 188 new secondary metabolites were summarized from 2015 to 2020 [7]. More than 200 cytotoxic or antitumor compounds isolated from marine Penicillium fungus were included from 1991 to 2017 [4]. Newly reported alkaloids produced by marine-derived Penicillium species were offered from 2014 to 2018 [8]. Recently, some remarkable achievements have been made in the study of marine-derived Penicillium fungi, including an MS/MS targeted molecular networking approach for the discovery of rare communesins [9], the investigation of the apoptosis mechanism of dicitrinone G from by the analyses of the protein–protein interaction (PPI) network and Western blot [10], and so on. These effective approaches have given rise to the generation of unique chemicals and huge biological diversity, making marine-derived Penicillium fungi a hotspot for utilization in the discovery of new drug leads.
A systematic review of the origins, structures, and bioactivities of 452 new NPs produced by marine-derived Penicillium species from January 2021 to December 2023 is provided in this review, based on 115 studies searching in the SciFinder database with marine-derived Penicillium as the key word, with English as the language. The review also covers fifty-one new marine natural products only described based on the HPLC-MS/MS analyses [9]; two previously reported marine natural products with significant new bioactivities [11,12], three known marine natural products supplied the structural NMR data [13,14,15], and two compounds were presented as new natural products [15,16], but their structures are not shown. In addition, due to the narrow publication timespan, seven pairs of new compounds possessing different structures were given the same trivial name, respectively. In this review, the first reported compounds were given the suffix a [17,18,19], and the other ones were given the suffix b [20,21].

2. New Bioactive Compounds from Marine-Derived Penicillium Fungi

2.1. Polyketides

2.1.1. Azaphilones

Azaphilones are a class of structurally diverse fungal metabolites that are mainly defined as polyketides possessing a highly oxygenated pyranoquinone bicyclic core and a quaternary carbon center [17]. A series of azaphilones with novel structures and remarkable bioactivities were reported from marine-derived Penicillium fungi. Penicil-azaphilones Ia–N (1, 2 and 69), epi-geumsanol D (3) and penidioxolanes C (4) and D (5) (Figure 1) were isolated from the sponge-derived P. sclerotiorum E23Y-1A culture [17]. Penicil-azaphilone Ia-N 9 showed moderate anti-inflammatory activity with an IC50 value of 22.63 ± 2.95 μM, whereas 4 exhibited various cytotoxic activities. The same strain produced two chlorinated azaphilones, penicilazaphilones F (10) and G (11) (Figure 1), with a moderate anti-inflammatory effect [22]. Based on a one strain–many compounds (OSMAC) approach, two new brominated analogs, 5-bromoisorotiorin (12) and penicilazaphilone Ha (13) (Figure 1), were obtained from P. sclerotiorum E23Y-1A by the addition of NaBr into the culture medium. Both showed moderate antibacterial activities against Staphylococcus aureus ATCC 25923 with inhibition zone diameters of 8.08 ± 0.01 and 7.50 ± 0.05 mm, respectively [18]. New azaphilones, penicilazaphilones Hb–Ib (1415), 11-epi-geumsanols B and F (1716), 8a-epi-hypocrellone A (18), and 8a-epi-eupenicilazaphilone C (19) (Figure 1), were isolated from algae-derived P. sclerotiorum [20,23]. Azaphilone 19 significantly promoted SMAD-mediated transcriptional activities stimulated by TGF-β [23]. Azaphilone E/Z isomers isochromophilone H (20a/b), sclerotiorins A (21) and B (22), ochlephilone (23), isochromophilone IV (24), and isochromophilone J (25a/b) (Figure 1) were isolated from the culture broth of the mangrove-derived fungus P. sclerotiorum HY5. Azaphilones 22 and 23 exhibited potent phytotoxicity against the growth of radicles and plumules on Amaranthus retroflexus L., with EC50 values ranging from 234.87 to 320.84 μM, compared to the positive control glufosinate-ammonium, with EC50 values of 555.11 μM for radicles, and 656.04 μM for plumules [24]. Chermesinones D–G (2629) (Figure 1) were isolated from marine-derived P. chermesinum FS625 [25]. Daldinins G-H (3031) (Figure 1) were isolated from the soft-coral-derived P. glabrum glmu 003 [26].

2.1.2. Isocoumarins

Penicillols A (32) and B (33) (Figure 1) featuring spiroketal rings were isolated from the mangrove-derived Penicillium sp. BJR-P2. Pannicillol B (33) exhibited significant inhibitory activity on NO production with an IC50 value of 12 μM [27]. Peniciisocoumarins I (34) and J (35) (Figure 1) were obtained from the mangrove-derived Penicillium sp. GXIMD 03001 [28].

2.1.3. Chromones

The marine-derived P. citrinum BCRC 09F458 yielded a class of rare chromone derivatives, epiremisporines C–H (3641) (Figure 1). Epiremisporines 37 and 38 could significantly inhibit fMLP-induced superoxide anion generation, with IC50 values ≤ 8.28 μM. Through the mitochondrial- and caspase 3-dependent pathways, 38 and 41 markedly induced the apoptosis of A549 with IC50 values of 43.82 ± 6.33 and 31.43 ± 3.01 μM, respectively. Furthermore, 41 obviously induced apoptosis of HT-29 cells, via Bcl-2, Bax, and caspase 3 signaling cascades [29,30]. Eleven 5,7-dioxygenated chromones penithochromones M–W (4252) (Figure 1), bearing an aliphatic acid side chain, were isolated from the deep-sea-sediment-derived P. thomii YPGA3 [31,32]. Penithochromones 4749 exhibited remarkable inhibition against α-glucosidase with IC50 values ranging from 842 to 1017 μM, which are more active than the positive control acarbose [31].

2.1.4. Citrinins

Citrinins, as an important class of polyketide mycotoxins, usually have core structure skeletons like benzopyran, benzofuran, and quinone-pyran, etc. [33]. Three extremely rare nitrogen-containing citrinin derivatives, isoquinocitrinins B–D (5355), and their corresponding enantiomers (53a/b, 54a/b, 55a/b) (Figure 2) were acquired from the hydrothermal-vent-sediment-derived Penicillium sp. TW131-64. These products exhibited potential anti-H. pylori activities towards the standard strain and multidrug-resistant clinical isolates with MIC values ranging from 0.25 to 8 μg/mL, indicating a comparable or even better killing activity than metronidazole. Citrinin derivatives (3R,4S)-8-hydroxy-6-methoxy-3,4,5-trimethylisochromane-7-carboxylatemethyl (56), (3R,4S)-6-hydroxy-8-methoxy -3,4,5-trimethylisochromane-7-carboxy latemethyl (57), and penicitrinone J (58) (Figure 2) were also obtained from this strain [33]. Rare carbon-bridged citrinin dimers, dicitrinones G–J (5962) (Figure 2) were isolated from starfish-derived Penicillium sp. GGF 16-1-2 and exhibited strong antifungal activities against Colletotrichum gloeosporioides with LD50 values ranging from 9.58 μg/mL to 16.14 μg/mL. Furthermore, 59 showed significant cytotoxicity against human pancreatic cancer cell lines BXPC-3 and PANC-1, which could induce apoptosis by activating caspase 3 proteins (CASP3) [10]. Neotricitrinols A–C (6365) (Figure 2), isolated from the marine-sediment-derived P. citrinum W23, feature a unique octacyclic carbon scaffold among the few reported citrinin trimers. Neotricitrinol 64 showed potential anti-osteoporosis activity by promoting osteoblastogenesis and inhibiting adipogenic differentiation on primary bone mesenchymal stem cells [34]. Xerucitrinins B and C (6667) (Figure 2) bearing a 6,6-spiroketal moiety were isolated from hydrothermal vent-associated P. citrinum Y34 [35]. Penicitrinol P (68) and dicitrinol D (69) (Figure 2) were isolated from the sponge-derived Penicillium sp. SCSIO 41302 [36] and Penicillium sp. SCSIO41303, respectively [37].

2.1.5. β-Resorcylic Acid

Resorcylic acid lactones (RALs) are structurally diverse polyketides, which usually consist of condensed resorcylic and macrolide cycles, sometimes possessing an open macrolide cycle [38]. Five β-resorcylic acid derivatives, 14-hydroxyasperentin B (70), β-resoantarctines A–C (7173) (Figure 2) and 8-dehydro-β-resoantarctine A (74) (Figure 2), were isolated from the brown-alga-derived P. antarcticum KMM 4685. β-resorcylic acid derivatives 7172 and 74 (Figure 2) inhibited the activity of p-glycoprotein at their noncytotoxic concentrations and consequently synergized with docetaxel in p-glycoprotein-overexpressing drug-resistant cells [38].

2.1.6. Verrucosidin

Verrucosidins belong to a family of highly reduced polyketides, generally sharing a methylated α-pyrone, a conjugated polyene linker, and an epoxidated tetrahydrofuran ring [39]. A pair of epimers, 9-O-ethylpenicyrones A (75) and B (76) (Figure 2), were isolated and identified from the marine-sediment-derived P. cyclopium SD-413. Epimers 75 and 76 showed antibiotic activity against aquatic pathogen A. hydrophilia, each with an MIC value of 8 μg/mL [40]. Poloncosidins A–K (7787) (Figure 2) [39,41] were identified from the cold-seep-sediment-derived P. polonicum CS-252. Poloncosidins 7786 were the first verrucosidins with a 2,5-dihydrofuran ring. Most of these compounds exhibited inhibitory activities against several human and aquatic pathogens with MIC values ranging from 4 to 32 μg/mL. Verrucosidinols A (88) and B (89) (Figure 2) were isolated from the marine-sediment-derived P. griseofulvum MCCC 3A00225 [42].

2.1.7. Citreoviridins

Citreoviridins H (90) and I (91) (Figure 2) were isolated from the mangrove-derived Penicillium sp. BJR-P2 [27]. Citreoviridins J-O (9297) (Figure 2) belonged to diastereomers of 6,7-epoxycitreoviridin with different chiral centers at C-2–C-7 and were isolated from the deep-sea-sediment-derived P. citreonigrum MCCC 3A00169 [43].

2.1.8. Nitrogen-Containing Polyketides

Fungal polyketide–amino acid hybrids are a large family of secondary metabolites produced by PKS-NRPS assembly lines [44]. The derivatives oxopyrrolidine A (98) and B (99) (Figure 2) were isolated based on bioactivity screening and chemical profiles from the marine-derived P. oxalicum MEFC104 [44]. 7-hydroxy-3,10-dehydrocyclopeptine (100) (Figure 2) was isolated from the mangrove-sediment-derived P. polonicum MCCC3A 00951 [45]. Fusarin derivatives steckfusarins A–E (101105) (Figure 2) were isolated from the green-algae-derived P. steckii SCSIO41040. Steckfusarin A (101) exhibited antioxidant activity against DPPH, with an IC50 value of 74.5 μg/mL [46].

2.1.9. Sorbicillinoids

Bisorbicillchaetones A-C (106108) (Figure 3) were the first examples of hybrid sorbicillinoids containing a coniochaetone unit and isolated from the deep-sea-sediment-derived Penicillium sp. SCSIO06868. Bisorbicillchaetones 106 and 107 exhibited moderate inhibitory effects on NO production in lipopolysaccharide (LPS)-activated RAW264.7 cells with IC50 values of 80.3 ± 3.6 μM and 38.4 ± 3.3 μM, respectively [47]. Various sorbicillinoids, including two hybrid sorbicillinoids, 10-methylsorbiterrin A (109) and dihydrotrichodermolidic acid (113); three bisorbicillinoids, epitetrahydrotrichodimer ether (110), demethyldihydro-trichodimerol (111) and bisorbicillpyrone A (112); and three monomeric sorbicillinoids, 5-hydroxy-dihydrodemethlsorbicillin (114), sorbicillpyrone A (115) and 5,6-dihydrovert-inolide (116) (Figure 3); were isolated from the deep-sea-sediment-derived Penicillium sp. SCSIO06871. Monomeric sorbicillinoid 114 displayed more potent inhibitory activity against α-glycosidase than acarbose with an IC50 value of 36.0 μM [48]. Sorbicatechols C (117) and D (118) (Figure 3) were isolated from deep-sea-derived P. allii-sativi MCCC3A00580. Sorbicatechol D (118) inhibited HT-29 cells in a dose-dependent manner [49]. A sorbicillinoid, (4E)-1-(4,6-dihydroxy-5-methylpyridin-3-yl)hex-4-en-1-one (119) (Figure 3), was isolated from the mangrove-derived Penicillium sp. DM815 [50].

2.1.10. Isochromans

Penicisteckins A–F (120125) (Figure 3) represented novel biaryl scaffolds containing both central and axial chirality elements and isolated from the beach-mud-derived P. steckii HNNU-5B18 [51].

2.1.11. α-Pyrone Polyketides

Six α-pyrone polyketides, penipyrols C–G (126130) (Figure 3) and methyl-penipyrol A (131), were isolated from the mangrove-derived Penicillium sp. HDN-11-131. Penipyrols 127129 possess a rare skeleton featuring γ-butyrolactone linked to an α-pyrone ring through a double bond. Penipyrol 127 can induce pancreatic β-cell regeneration in zebrafish at 10 μM, demonstrating promising anti-diabetes potential [52].

2.1.12. Hirsutellones

The natural hirsutellones are made of a decahydrofluorene polyketide core (rings A, B and C) involved in a highly strained 12- or 13-membered para-cyclophane (ring D) and highly functionalized 5-hydroxypyrrolidinone [53]. Perpyrrospirone A (132) (Figure 3) was the first example of hirsutellone peroxide from the marine-derived P. citrinum, and characterized an unprecedented 6/5/6/8/5/13/6 oxahexacyclic scaffold with a unique peroxide-bridged 8,9-dioxa-2-azaspiro[4.7] dodecane core [53].

2.1.13. Xanthones and Benzophenones

Xanthones, known as 9H-xanthen-9-ones, are a class of yellow compounds bearing a dibenzo-γ-pyrone scaffold, which are often regarded as privileged structures for binding with a variety of targets [54]. Tetrahydroxanthone 11-O-acetylaspergillusone B (133) and the fully aromatic xanthone 7-dehydroxyhuperxanthone A (134) (Figure 3) were isolated from the deep-sea-sediment-derived Penicillium sp. MCCC 3A00126 [54]. Penicixanthene E (135) (Figure 3), the first reported xanthene derivative in which a carbon–carbon double bond was reduced, was isolated from the mangrove-derived Penicillium sp. GXIMD 03101 [55]. Benzophenone derivative penibenzophenone C (136) (Figure 3) and a new natural product, penibenzophenone D, were isolated from the mangrove-derived Penicillium sp. Penibenzophenone C (136) showed moderate antibacterial activity against methicillin-resistant S. aureus with an MIC value of 3.12 μg/mL [16].

2.1.14. Hydroxybenzenes

Peniketide A (137) and a methyl ester of penipyrol A (138) (Figure 3) were isolated from the marine-sediment-derived Penicillium sp. SCZ-1. Peniketide A 137 bearing a two-carbon side chain at C-2 is seldom found among natural isocoumarins [56]. The deep-sea-sediment-derived P. citrinum W17 yielded penidihydrocitrinins A–C (139141) (Figure 3). Three isolates exhibited significant inhibitory effects on LPS-stimulated nitric oxide (NO) production in murine brain microglial BV-2 cells in a dose–response manner [57]. Peniciphenalenin G (142) (Figure 3) was isolated from the marine-derived P. oxalicum [58]. Penicinone C (143) (Figure 3) was identified from the mangrove-derived Penicillium sp. LA032 [59]. Six new polyketide derivatives (144149) (Figure 3) were isolated from the hydrothermal-vent-sediment-derived Penicillium sp. TW58-16. Polyketide derivatives 144147 showed strong a-glucosidase inhibitory effects with inhibition rates of 73.2%, 55.6%, 74.4%, and 32.0%, respectively, which were comparable with or even better than that of acarbose, a known α-glucosidase inhibitor [60]. Coniochaetone N (150) (Figure 3) was isolated from the deep-sea-sediment-derived Penicillium sp. SCSIO06868 [61].

2.1.15. Lactones

Penicinones A (151) and B (152) (Figure 3) were identified from the mangrove-derived Penicillium sp. LA032. Penicinone A 151, a rare furo[3,4-b]pyran-5-one skeleton with an n-heptyl moiety, was identified and found to exhibit significantly cytotoxic activity against the HepG2 cells, with an IC50 value of 3.87 ± 0.74 μM [59]. Walterolactone E (153) (Figure 3) was isolated from the hydrothermal-vent-sediment-derived Penicillium sp. TW58-16 [62].

2.1.16. Olefinic Acids and Their Derivatives

Tanzawaic acids are a small class of polyketides, characterized by a trans-decalin (A/B fusion) scaffold, isolated mainly from the genus Penicillium [63]. The coral-derived P. steckii AS-324 yielded a series of tanzawaic acids, including steckwaic acids A–D (154157), 11-ketotanzawaic acid D (158), 6,15-dihydroxytanzawaic acid M (159), 15R-methoxy-tanzawaic acid M (160), 15S-methoxytanzawaic acid M (161), 8-hydroxytanzawaic acid M (162), and 8-hydroxytanzawaic acid B (163) [64], steckwaic acids Ea-Ia (164168), 18-O-acetyltanzawaic acid R (169), 10-hydroxytanzawaic acid U (170), and 13R-tanzawaic acid S (171) [19] (Figure 4). Among them, 171 showed potent activity against Escherichia coli with an MIC value of 8 μg/mL. Steckwaic acids Eb-Ib (172176) and J-K (177178) (Figure 4) were isolated from the green-algae-derived P. steckii SCSIO 41040. Steckwaic acid 173 inhibited LPS-induced nuclear factor kappa-B (NF-κB) with an IC50 value of 10.4 μM, which is the first report of osteoclastogenesis inhibitory activity for tanzawaic acid derivatives [63]. Penicisteck acids A–D (179182) (Figure 4) were isolated from the mangrove-derived P. steckii SCSIO 41025. Penicisteck acids 179181 were highly oxygenated decalin derivatives harboring an unusual propanoic acid unit at C-1 [65]. Penifellutins A (183) and B (184), possessing a 22 carbons linear skeleton, were isolated from the co-culture of the deep-sea-derived fungi P. crustosum PRB-2 and P. fellutanum HDN14-323 along with two esterification products, penifellutins C (185) and D (186) (Figure 4). Penifellutins A (183) and B (184) showed obvious inhibitory activity on the liver hyperplasia of zebrafish larvae at a concentration of 10 μmol/L, while penifellutins C (185) and D (186) showed no activity, indicating that two carboxyls in the structure were important active sites [66].

2.1.17. Other Polyketides

Rubenpolyketone A (187) (Figure 4) featuring cyclohexenone condensed with a methyl octenone chain was identified from the Magellan Seamount-derived P. rubens AS-130 [67]. Oxalichroman A (188) and oxalihexane A (189) (Figure 4) were isolated from the red algae-derived P. oxalicum 2021CDF-3. Oxalihexane A (189), formed by a cyclohexane and cyclohexanone moiety via an ether bond, showed a remarkable inhibitory effect on the human pancreatic cancer PATU8988T cell line through downregulation of the expression level of cyclin D1 [68]. Leptosphaerone D (190) (Figure 4) was isolated from the hydrothermal-vent-sediment-derived Penicillium sp. TW58-16 [62]. 15-O-methyl ML-236A (191) (Figure 4) was isolated from the deep-sea-sediment-derived P. solitum MCCC 3A00215 [69].

2.2. Alkaloids

2.2.1. Indoles

Communesins are a class of complex indole alkaloids isolated from the Penicillium fungi. The marine-sediment-derived P. expansum was studied using a targeted molecular networking approach, allowing the detection of 55 new communesins. Among of them, communesins M-P (192195) (Figure 5) were isolated and showed moderate cytotoxicity against KB and MCF-7 human cancer cell lines in comparison to the positive control docetaxel [9]. The coral-derived P. dimorphosporum KMM 4689 yielded the very first deoxyisoaustamide alkaloid deoxy-14,15-dehydroisoaustamide (196) [70] and seven deoxyisoaustamide derivatives (197203) [71] (Figure 5). Deoxyisoaustamide derivatives 199, 201 and 202 (Figure 5) showed a statistical increase in paraquat-treated Neuro-2a cell viability by 30–39% at a concentration of 1 μM. Penilline D (204) (Figure 5) was isolated from the Antarctic fungus Penicillium sp. SCSIO 05705 [72]. Penindolacid A (205) (Figure 5) was isolated from the marine-sediment-derived Penicillium sp. LW92 [73]. Prenylated indole diketopiperazine alkaloids (PIDAs) penicamides A (206) and B (207) (Figure 5) were identified from the mangrove-derived Penicillium sp. LA032. Penicamide A (206) was the first example of PIDAs featuring a 6/5/8/6/5 pentacyclic ring system with an α-hydroxy group at C-11[59]. 11S-(−)-penilloid A (208) and 11R,14E-(+)-penilloid A (209) (Figure 5) were isolated from the marine-mud-derived Penicillium sp. ZZ1750 [74].

2.2.2. Pyridones

Eleven new pyridone alkaloids, penicipyridones A-K (210220) (Figure 5), were isolated from the marine-derived P. oxalicum QDU1. Penicipyridones 210, 213214, 217 and 219220 exhibited moderate inhibitory effects on NO production in the LPS-induced RAW264.7 macrophages, with IC50 values ranging from 9.2 to 19 μM [75].

2.2.3. Quinolinones

Penicinolone (221) (Figure 5) was isolated from the sponge-derived Penicillium sp. SCSIO41033 [76]. AChE-inhibitory-activity-guided studies on the mangrove-derived P. citrinum YX-002 led to the isolation of quinolactone A (222), quinolactacin C1 (223), and 3-epi-quinolactacin C1 (224) (Figure 5). Quinolactone A (222) showed moderate AChE inhibitory activity with an IC50 value of 27.6 μmol/L [77]. N-methyl-4-quinolones quinolactacin E (225a/b), quinolactacins F1–F2 (226227) and quinolactacin G (228a/b) (Figure 5) were isolated from the sponge-derived Penicillium sp. SCSIO 41303. Quinolactacin 226 exhibited enzyme inhibition activity against PL with an IC50 value of 24.6 μg/mL [37]. Four racemic mixtures, (±)-oxypenicinolines A–D (229232), along with penicinolines F (233) and G (234) (Figure 5) were isolated from the mangrove-derived P. steckii SCSIO 41025. Racemic mixtures 229232 shared an unusual 6/6/5/5 tetracyclic system incorporating a rare tetrahydro-pyrrolyl moiety, while 229 displayed α-glucosidase inhibitory activity with an IC50 value of 317.8 μM, which was more potent than that of acarbose (461.0 μM) [78].

2.2.4. Decahydrofluorene-Class Alkaloids

Pyrrospirones K–Q (235241) (Figure 5) were isolated from the soft-coral-derived Penicillium sp. SCSIO 41512. Pyrrospirones 235 and 237 possessed a novel decahydrofluorene-class alkaloid skeleton with 6/5/6/8/5/6/13 and 6/5/6/5/6/13 polycyclic systems, respectively. Pyrrospirones 235237 and 239 showed antibacterial activity against all or some of the six pathogens B. amyloliquefaciens, B. subtilis, E. coli, S. aureus, S. aureus MRSA, and S. agalactiae. Pyrrospirones 238 and 240 displayed mild inhibitory activity against several PTPs with IC50 values of 39.4–100 μM [79].

2.2.5. Piperazines

A trithiodiketopiperazine derivative, adametizine C (242) (Figure 5), was isolated from the mangrove-sediment-derived P. ludwigii SCSIO 41408. Adametizine C (242) showed cytotoxicity against prostate cancer cell line 22Rv1 with an IC50 value of 13.9 μM, and the strongest inhibitory activity against RANKL-induced osteoclast differentiation in bone marrow macrophage cells with 10 μM [80]. A diketopiperazine alkaloid, (8S,9R,12R,18S)-12-hydroxyl-fumitremorgin B (243) (Figure 5), was isolated from the hydrothermal-vent-sediment-derived Penicillium sp. TW58-16 [62]. Three epithiodiketopiperazine alkaloids, penigainamides A–C (244246) (Figure 5), were isolated from the marine-derived P. steckii YE [81].

2.2.6. Tetramic-Acid-Based Alkaloids

Tolypocladenols D-F (247249) (Figure 6) were isolated from the fresh and healthy leaves of the Apocynum venetum-derived fungus P. oxalicum QDU1. Tolypocladenol 249 exhibited moderate inhibitory effects on NO production in the LPS-induced RAW264.7 macrophages, with an IC50 value of 14 ± 1 μM [75]. Penicillenols G1–G2 (250251) and H (252) (Figure 6) were isolated from cultures of the deep-sea-sediment-derived Penicillium sp. SCSIO06868. Penicillenol H (252) exhibited potent inhibitory activities against S. aureus and methicillin-resistant S. aureus with MIC values of both 2.5 mg/mL [61].

2.2.7. Amines and Amides

(Z)-4-(5-acetoxy-N-hydroxy-3-methylpent-2-enamido) butanoate (253) (Figure 6) was isolated from the mangrove-derived P. oxalicum HLLG-13 and showed significant growth inhibition activities against newly hatched Helicoverpa armigera Hubner larvae, with an IC50 value of 200 μg/mL [15]. Polonimides E (254) and D (255) (Figure 6) were isolated from the sponge-derived Penicillium sp. SCSIO 41413 [82]. Speradine I (256) (Figure 6) was isolated from the soft-coral-derived Penicillium sp. SCSIO 41038 [83]. (S)-2-acetamido-4-(2-(methylamino)phenyl)-4-oxobutanoic acid (257) (Figure 6) was isolated from the deep-sea-gammarid-shrimp-derived P. citrinum XIA-16 [84]. A pentacyclic alkaloid, citrinadin C (258) (Figure 6), was isolated from the deep-sea-sediment-derived P. citrinum and showed cytotoxic activity against human liver cancer cell line MHCC97H, with an IC50 value of 16.7 μM [85]. (2S,2′R,3R,3′E,4E,8E)-N-2′-hydroxyhexadecanoyl-2-amino -9-methyl-4,8-octadecadiene-1,3-diol (259) (Figure 6), a ceramide, was isolated from the seawater-derived P. chrysogenum Y20-2. (2S,2′R,3R,3′E,4E,8E)-N-2′-hydroxyhexadecanoyl-2-amino -9-methyl-4,8-octadecadiene-1,3-diol (259) showed no pro-angiogenic activity using a zebrafish model [86]. N-acetyl-d-glucosamines penichryfurans A (260) and B (261) (Figure 6) were isolated from the red-alga-derived P. chrysogenum. Penichryfuran A (260) exhibited strong cytotoxicity against the HepG2 cell line with an IC50 value of 9.0 μM [87]. Talaroenamines F1–F19 (262280) (Figure 6) were isolated from the wetland-derived P. malacosphaerulum HPU-J01 using a one-pot/two-stage precursor-directed biosynthesis approach. Talaroenamine 275 was cytotoxic against the K562 cell line with an IC50 value of 2.2 μM [88]. Peniokaramine (281) and penipyranopyridine (282) (Figure 6) were isolated from the hydrothermal-vent-sediment-derived Penicillium sp. LSH-3-1. Peniokaramine (281) showed moderate cytotoxic activity against A549 cells with an inhibition percentage of 53.43 ± 5.89% [89]. Penicidihydropyridones A (283) and B (284) (Figure 6) were isolated from the sponge-derived Penicillium sp. B9. Both of them intriguingly appeared to perturb PD-L1/PD-1 interactions with a considerable inhibitory rate of 88.40% for 283 and 70.72% for 284 with a concentration of 10 μg/mL [90]. (+)-solitumidine D (285) and (±)-solitumidine E (286) were isolated from the marine-sediment-derived P. solitum MCCC 3A00215 [69]. Penicmariae-crucis C acid (287), N-(6-hydroxy-2-oxoindolin-3-ylidene)-5′-methoxy-5′-oxobutyl-amine oxide (288), and methyl-1′-(N-hydroxyacetamido)-butanoate (289) (Figure 6) were isolated from the mangrove-derived P. steckii SCSIO 41025 [65]. Penigrisamide (290), aurantiomoate C (291), N,N-pyroglutamylleucinmethylester (292), methyl-2S-hydroxy-3-methylbutanoyl-L-leucinate (293), and 6R,7-dihydroxy-3,7-dimethyloctanamide (294) (Figure 6) were isolated from the marine-sediment-derived P. griseofulvum MCCC 3A00225 [42].

2.2.8. Other Alkaloids

Sulfoxanthocillin (295) (Figure 6) was isolated from the deep-sea-sediment-derived Penicillium sp. SCSIO sof101. Sulfoxanthocillin (295) showed significant activity against series pathogens with MIC values ranging from 0.06 to 8.0 μg/mL and relatively low cytotoxicity against human tumor cell lines [91]. Penipyridinone B (296) (Figure 6) was isolated from the marine-mud-derived Penicillium sp. ZZ1750. Penipyridinone B (296) represented the first example of its structural type and showed potent antiglioma activity, with IC50 values of 2.45 μM for U87MG cells and 11.40 μM for U251 cells [74].

2.3. Terpenoids

2.3.1. Sesquiterpenes

A linear sesquiterpenoid, chermesiterpenoid D (297) (Figure 7), was identified from the Magellan Seamount-derived P. rubens AS-130 [67]. A series of eremophilane-type sesquiterpenes, copteremophilanes A–J (298307) (Figure 7), were isolated from the marine-sponge-derived P. copticola. Analogs 298, 299, and 307 represented a group of uncommon skeletons of eremophilanes with an aromatic ring and a methyl migration from C-5 to C-9. The incorporation of a chlorinated phenylacetic unit in 300306 has rarely been found in nature; 304 showed a neuroprotective effect through increasing the viability of A25-35-induced PC12 cells, whereas 305 exhibited selective inhibition against A549 with an IC50 value of 3.2 ± 0.1 μM [92]. A drimane sesquiterpenoid, astellolide Q (308) (Figure 7), was isolated from the mangrove-soil-derived Penicillium sp. N-5, combined with compound V [14]. A drimane sesquiterpene ester, chrysoride A (309) (Figure 7), was isolated from the red-alga-derived P. chrysogenum LD-201810 and showed moderate cytotoxicity against HepG2 and HeLa cancer cell lines, with IC50 values of 28.9 and 35.6 μM, respectively [93]. The marine-derived Penicillium sp. ZZ1283 yielded a drimane sesquiterpene, lactone purpuride D (310) (Figure 7), which significantly inhibited the growth of methicillin-resistant S. aureus, E. coli and C. albicans with MIC values of 4, 3 and 8 μg/mL, respectively [94]. Acorane-type sesquiterpenes feature a spiro[4.5]decane core with an isopropyl unit at C-1 and dimethyl substitution at C-4 and C-8, which markedly differs from other types of the sesquiterpene family [95]. Eighteen acorane-type sesquiterpenes, bilaiaeacorenols A–R (311328) (Figure 7), were identified from the deep-sea-sediment-derived P. bilaiae F-28. Sesquiterpene 328 exhibited efficient reduction against NO production in LPS-induced BV-2 macrophages in a dose-dependent manner, and it abolished LPS-induced NF-κB in the nucleus of BV-2 microglial cells, along with the inhibition of iNOS and COX-2 at a cellular level [95]. Citreobenzofurans D–F (329331) and phomenones A–B (332333) (Figure 7) were isolated from the mangrove-derived Penicillium sp. HDN13-494. Citreobenzofurans 330 and 331 are eremophilane-type sesquiterpenoids with rare benzofuran frameworks. Phomenone B (332) contained a rare thiomethyl group, which was the first report of this kind of sesquiterpene with sulfur elements in the skeleton; 333 showed moderate activity against Bacillus subtilis, with an MIC value of 6.25 μM [96]. (2S,3S,5S,6S,7S,8R,11S,12R)-15-deacetyl-7,8-dihydroxycalonectrin (334) and 1-methyl-4-[3,4,5-trihydroxy-1,2,2-trimethylcyclopently] benzene (335) (Figure 7) were isolated from the deep-sea-derived Penicillium sp. LXY140-R. 1-methyl-4-[3,4,5-trihydroxy-1,2,2-trimethylcyclopently] benzene (335) showed potent antiproliferative activity against HCT-116 cell lines with an IC50 value of 124.12 μM [97]. Nor-bisabolane derivative enantiomers (±)-1 (336a/b) (Figure 7) were isolated from the algal-derived P. chrysogenum LD-201810 [98]. Two drimane sesquiterpenes, (4S,5R,9S,10R)-11,13-dihydroxy-drim-7-en-6-one (337) and (4S, 5R,9S,10R)-11-hydroxy-13-carboxy-drim-7-en-6-one (338) (Figure 7), were isolated from the hydrothermal-vent-sediment-derived Penicillium sp. TW58-16. (4S,5R,9S,10R)-11,13-dihydroxy-drim-7-en-6-one (337) showed a strong a-glucosidase inhibitory effect with an inhibition rate of 35.4% [60].

2.3.2. Diterpenes

Resistance has been found in many clinical anti-influenza A virus (IAV) drugs. Therefore, developing a safe and effective agent with a unique structure is urgently needed to combat IAV infection [99]. During the search for anti-IAV marine natural products, a series of new indole-diterpenoids have been isolated. Penijanthine E (339) (Figure 7), obtained from the marine-derived P. citrinum ZSS-9, showed antiviral activity against IAV of A/WSN/33(H1N1) and A/PR/8/34(H1N1) strains with IC50 values of 12.6 and 18.9 μM, respectively [99]. The marine-derived P. janthinellium co-cultured with Paecilomyces formosus led to the isolation of janthinellumines A–I (340348). Janthinellumines 340, 341 and 346 (Figure 7) displayed significant activities against two strains of A/WSN/33 (H1N1) and A/Hong Kong/1/68 (H3N2) with IC50 values of 3.8 and 13.3 μM, respectively, stronger than those of the positive control T-705. Furthermore, the PTP inhibitory activity of 340341, 343344 and 348 had the best inhibitory activity towards PTP1B with IC50 values ranging from 0.6 to 9.2 μM, most of which were stronger than that of the positive control Na3VO4 (IC50 = 8.5 μM) [100]. Additionally, oxalierpenes A (349) and B (350) (Figure 7) were obtained from the mantis-shrimp-derived P. oxalicum. Oxalierpene A (349) represents the first indole-diterpenoid derivative with a five-membered ring of 4-hydroxy-5,5-dimethyl-dihydrofuran-3-one as a side chain. Oxalierpene B (350) had a unique 6/5/6/5/5/6/6/5/5 ring system. Oxalierpenes A (349) and B (350) showed antiviral activity against the H1N1 virus and respiratory syncytial virus (RSV), with IC50 values ranging from 2.8 to 9.4 μM [101]. 4-Hydroxyleptosphin C (351) and 13-epi-conidiogenone F (352) (Figure 7) were isolated from the marine-sediment-derived P. antarcticum KMM 4670. 4-Hydroxyleptosphin C (351) and 13-epi-conidiogenone F (352) inhibited C. albicans growth by 30.4% and 27.9% at 12.5 μM, respectively. Moreover, they significantly inhibited sortase A activity by 28.2% and 36.9% at 50 μM, respectively [102]. Shearinines R–T (353355) and 22-hydroxyshearinine I (356) (Figure 7) were isolated from mangrove-sediment-derived Penicillium sp. UJNMF0740 [103]. Conidiogenones J (357) and K (358) (Figure 7) were isolated from the mangrove-derived P. oxalicum HLLG-13. Both of them showed significant growth inhibition activities against newly hatched Helicoverpa armigera Hubner larvae, with IC50 values of 200 μg/mL [15]. Penerpenes K–N (359362) (Figure 7) were isolated from the bivalve mollusk-derived Penicillium sp. KFD28 [104]. Epipaxilline (363) and penerpene J (364) (Figure 7) were isolated from the marine-derived Penicillium sp. KFD28. Epipaxilline (363) and penerpene J (364) showed inhibitory activities against PTP1B with IC50 values of 31.5 and 9.5 μM, respectively. Penerpene J (364) also showed inhibitory activities against TCPTP with an IC50 value of 14.7 μM [105].

2.3.3. Meroterpenes

Meroterpenoids are a family of hybrid natural products with high scaffold diversity and significant pharmacological activities [106]. Peniscmeroterpenoids H–N (365371) (Figure 8) were isolated from the marine-derived P. sclerotiorum GZU-XW032. Among them, 365 featured a unique 2-oxaspiro[5.5] undeca-4,7-dien-3-one motif. Peniscmeroterpenoids 366 and 367 owned rare 6(D)/5(E) fused rings. Peniscmeroterpenoid 368 was the first case where the C-24 was oxidized. Peniscmeroterpenoid 369 exhibited a moderate inhibitory effect in NO production with an IC50 value of 48.04 ± 2.51 μM [106]. Andrastin I (372) (Figure 8) was isolated from the seafloor-sand-derived P. ochrochloron [107]. Chermesins E–H (373376) (Figure 8) were isolated from the alga-derived P. chermesinum EN-480. Chermesin 373 showed effective activities against the aquatic pathogens E. tarda and V. anguillarum with MIC values of 0.5 μg/mL, respectively. Chermesin 374 showed powerful activities against human pathogenic bacterium E. coli with an MIC value of 1 μg/mL [108]. Seven meroterpenoids, peniscmeroterpenoids A–G (377383) (Figure 8), were isolated from the marine-derived P. sclerotiorum GZU-XW03-2. Peniscmeroterpenoid 377 possessed an unprecedented and highly oxidized 6/7/6/5/5 pentacyclic system, featuring a unique tetrahydrofuro [2,3-b]furan-2(3H)-one motif. Peniscmeroterpenoids 378381 with 6(D)/5(E) fused rings were rare in natural products, and 381 was the first example of a berkeleyone analogue stripped of the methyl ester fragment. In bioassays, 377 and 380 inhibited the production of NO in RAW264.7 cells with IC50 values of 26.60 ± 1.15 and 8.79 ± 1.22 μM. Moreover, 380 significantly suppressed the production of pro-inflammatory mediators (COX-2, IL-1β and IL-6) and the protein expression of the enzyme iNOS [109].
Meroterpenthiazole A (384) [110] and nine andrastones, namely citrehybridonol B (385), andrastin G (386), and andrastones B–H (387393) [111] (Figure 8), were isolated from the deep-sea-derived P. allii-sativi MCCC 3A00580. Meroterpenthiazole A (384) had a rare benzothiazole moiety and significantly inhibited the transcriptional effect of retinoid X receptor (RXR)-α (KD = 12.3 μM). Citrehybridonol B (385) had a novel hemiketal moiety, and 386 was the first example to possess a novel tetrahydrofuran moiety via C-7 and C-15. Andrastones 391393 were the first three examples of andrastones bearing a doublet methyl (C-18) at C-16; 387 potently decreased degranulation with an IC50 value of 40.4 μM. Three andrastin-type meroterpenoids, hemiacetalmeroterpenoids A–C (394396) (Figure 8), were isolated from the mangrove-soil-derived Penicillium sp. N-5. Hemiacetalmeroterpenoid A (394) possessed a unique and highly congested 6,6,6,6,5,5-hexa-cyclic skeleton and exhibited significant antimicrobial activities against P. italicum and C. gloeosporioides with an MIC value of 6.25 μg/mL [14]. The other three andrastin-type meroterpenoids, penimeroterpenoids A–C (397399) (Figure 8), were isolated from the deep-water-sediment-derived Penicillium sp. Penimeroterpenoid A (397) showed moderate cytotoxicity against the A549, HCT116, and SW480 cell lines [112]. The unusual austins-type meroterpenoids penicianstinoids C–E (400402) (Figure 8) were obtained from the mangrove-derived Penicillium sp. TGM112. Penicianstinoid 400 owns two unusual spirocyclic moieties, 401 contains an octahydro-2H-chromen-2-one unit, and 402 has an uncommon five-membered ether ring. Penicianstinoids 400 and 402 inhibited the growth of newly hatched H. armigera Hubner larvae with IC50 values of 100 and 200 μg/mL, respectively [113].

2.4. Steroids

Rubensteroid A (403) (Figure 9) was isolated from the Magellan Seamount-derived P. rubens AS-130. Rubensteroid A (403) had a rare 6/6/6/6/5 pentacyclic system and exhibited strong antibacterial activity against E. coli and Vibrio parahaemolyticus, both with an MIC value of 0.5 μg/mL [114]. Andrastin H (404) (Figure 9) was isolated from the mangrove-derived P. oxalicum HLLG-13 and showed significant growth inhibition activity against newly hatched H. armigera Hubner larvae, with an IC50 value of 50 μg/mL [15]. A unique 6/6/6/6/5 steroid, solitumergosterol A (405) (Figure 9), was isolated from the deep-sea-sediment-derived P. solitum MCCC 3A00215 [115].

2.5. Peptides

Two linear peptides, penicamides A (406) and B (407) (Figure 9), were isolated from the soft-coral-derived Penicillium sp. SCSIO 41512 [21]. Penicillizine A (408) (Figure 9) was isolated from the from the Red Sea tunicate-derived P. commune DY004 [116].

2.6. Others

Penioxa acids A (409) and B (410) (Figure 10) were isolated from the marine-sediment-derived P. oxalicum BTBU20213011 [117], and (Z)-5-acetoxy-3-methylpent-2-enoic acid (411) (Figure 10) was isolated from the mangrove-derived P. oxalicum HLLG-13 with a new natural product (2-hydroxy-5-methoxyphenyl) methyl acetate [15]. Antaketide A (412) (Figure 10) was isolated from the marine-sediment-derived P. antarcticum KMM 4670 [102]. A butyrolactone congener ochrochloronic acid (413) (Figure 10) was yielded from the seafloor-sand-derived P. ochrochloron co-cultivating with Bacillus subtilis [107]. (Z)-4-((6,7-dihydroxy-3,7-dimethyloct-2-en-1-yl)oxy)benzoic acid (414) (Figure 10) was isolated from a marine-mud-derived P. arabicum ZH3-9 [118]. Three α-pyrone derivatives, annularins L–N (415417) (Figure 10), were isolated from the rhizospheric soil of the mangrove-derived P. herquei MA-370 [119]. Peniprenylphenol A (418) (Figure 10), a tetrasubstituted benzene derivative, was isolated from the mangrove-sediment-derived P. chrysogenum ZZ1151 and had antimicrobial activities against human pathogenic methicillin-resistant S. aureus, E. coli and C.albicans with MIC values of 6, 13, and 13 μg/mL, respectively [120]. 13-(11-Hydroxy-8-(4-hydroxy-1,6-dimethoxybenzyl)-9 -methoxy-12-methylphenyl) propan-15-one, a benzene derivate (419) (Figure 10), was isolated from the green-algae-derived P. steckii SCSIO 41040 [63]. A phloroglucinol derivative, speradine J (420) (Figure 10), was isolated from soft-coral-derived Penicillium sp. SCSIO 41038 [83]. A butenolide derivative, eutypoid F (421) (Figure 10), was isolated from the sponge-derived Penicillium sp. SCSIO 41413 and exhibited an inhibitory effect against the enzyme PI3K with an IC50 value of 1.7 μM [82]. Penisterines A (422) and C–E (423425) and penisterine A methyl ether (426) (Figure 10) were isolated from the brown-alga-derived P. sumatraense SC29. Penisterine E (425) was a unique 6/6/6-tricyclic ether with an acetal and two hemiketal functionalities. Among these, 424 inhibited human endothelial progenitor cell (EPC) growth, migration, and tube formation without any cytotoxic effect. Furthermore, in in vivo bioassays, the percentages of angiogenesis of 423 on Tg(fli1:EGFP) transgenic zebrafish were 54% and 37% as the treated concentration increased from 10.2 to 20.4 μg/mL, respectively. The percentages of angiogenesis of 424 were 52% and 41% as the treated concentration increased from 8.6 to 17.2 μg/mL, respectively [121]. Five alkane derivatives (427431) (Figure 10) were isolated from the mangrove-sediment-derived P. ludwigii SCSIO 41408. Alkane derivatives 429 and 431 exhibited obvious inhibitory activities against LPS-induced NF-kB with IC50 values of 10.7 and 21.5 μM, respectively. Moreover, in a further study of their effects on RANKL-induced osteoclastogenesis, alkane derivative 429 was found to be able to suppress the RANKL-induced osteoclast differentiation in BMMCs, with a concentration of 10 μM [80].
5,6-dihydroxy-3-methoxyhex-2-enoic acid (432) (Figure 10) was isolated from the deep-sea-sediment-derived Penicillium sp. LXY140-3 co-culturing with Penicillium sp. LXY140-R [97]. 6-acetyl-4-methoxy-3,5-dimethyl-2H-pyran-2-one (433) and (2E,4E)-5-((2S, 3S,4R,5R)-3,4-dihydroxy-2,4,5-trimethyltetrahydrofuran-2-yl)-2,4-dimethylpenta-2,4-dienal (434) (Figure 10) were identified from the mangrove-derived P. polonicum H175, and they showed no hypoglycemic effect by the Tg (Ins: htBidTE-ON; LR) zebrafish [122]. 5-glycopenostatins F (435) and I (436) (Figure 10), characterized by an unprecedented PKS scaffold bearing a glucose unit, were isolated from the sponge-derived P. Copticola. Their activities have not been identified [92]. (±)-Tetraketides 437a/b were isolated from the sponge-derived Penicillium sp. SCSIO 41302. (-)-Tetraketide 437b exhibited significant inhibitory activities against pancreatic lipase and acetyl cholinesterase with an IC50 value of 48.5 μM, which indicated that different chiral centers between enantiomers (437a/b) may result in different biological activities (IC50 value of 437 against PL > 100 μg/mL) [36]. 8-hydroxyhelvafuranone (438), methyl-3,7,9-trihydroxydecanate (439), and 9-hydroxy-3,7-epoxydecanoic acid (440) (Figure 10) were isolated from the marine-sediment-derived P. griseofulvum MCCC 3A00225 [42]. Phthalides chrysoalides A (441) and B (442) (Figure 10) were isolated from the red-alga-derived P. chrysogenum LD-201810 [98]. P-terphenyl derivatives peniterphenyls A–C (443445) (Figure 10) were obtained from the deep-sea-sediment-derived Penicillium sp. SCSIO 41030. Peniterphenyl 444 represents the first reported natural product possessing a 4,5-diphenyl-substituted 2-pyrone derivative. Peniterphenyls 443 and 444 significantly increased the viability of Vero cells infected with HSV-1/2 with the EC50 values of the p-terphenyls ranging from 1.4 ± 0.6 to 9.3 ± 3.7 μM, with 50% cytotoxicity concentration values greater than 100 μM against Vero cells [123].
The names and numbers of all new compounds according to their classes, the sources from which marine-derived Penicillium were isolated, the biological activities of new compounds, and corresponding references are listed in Table 1.

3. Statistical Analysis of New Natural Products from Marine-Derived Penicillium

Penicillium fungi can establish a good relationship with different marine organisms and marine environments. According to the statistical results, sediments and mangroves were the main sources or hosts of marine-derived Penicillium fungi for producing new natural products, nearly 56% (Figure 11).
The new natural products had diverse chemical structures including polyketides, alkaloids, terpenoids, steroids, peptides, and others. Figure 12 shows the proportion of new bioactive compounds in each chemical class. A total of 194, 107 and 107 new compounds belong to polyketides, alkaloids and terpenoids, respectively, adding up to more than 90% of the total. Similarly, these three classes contribute 94% of all new bioactive compounds. The highest proportion of bioactives belongs to the largest number of polyketides (86) with 44.3%, followed by terpenoids (41) with 39.3% and alkaloids (42) with 38.3%.
The new compounds were counted only once when they were analyzed for bioactivity or inactivity. If the article did not provide a description of strong, moderate or weak activity of bioactive compounds, we gave this description according to bioactivity potency criteria used in the review [124]. The multi-active compounds were counted multiple times when they were classified according to anti-cancer/cytotoxicity, anti-inflammatory, antibacterial, antifungal, antiviral, enzyme inhibitory, antioxidant, and anti-allergy activities, as well as others [125].
Figure 13 shows the percentage distribution of new compounds with different bioactivities for 2021–2023. Among them, 24.3% of the new bioactive compounds showed antibacterial activity with the number of 44. This was followed by cytotoxic activity at 38 (21%), enzyme inhibition activity at 30 (16.6%), and anti-inflammatory activity at 27 (14.9%).
Figure 14A shows the proportion distribution of new compounds with different bioactivities in each chemical class for 2021–2023; peptides are not listed due to the absence of activity results. Polyketides displayed antibacterial activity as the dominant activity with a proportion of 29%, highlighting that they encompass many potential antibacterial drug leads. For alkaloids, cytotoxic compounds accounted for 31.7% of the total active compounds, while terpenoids displayed relatively high enzyme inhibitory with a proportion of 21.4%. Figure 14B shows the proportion distribution of new compounds with different chemical classes in each bioactivity for 2021–2023. The major contributors to antibacterial activity are polyketides. The most promising anti-cancer/cytotoxicity agents from marine-derived Penicillium fungi appear to be alkaloids. The main anti-inflammatory and enzyme inhibitory metabolites are still polyketides.
It should be noted that not all new metabolites isolated from the marine-derived Penicillium fungi were tested for biological activity because of scarcity of quantity [37,52,59,79,92], while many bioactive compounds were only studied for one type of bioassay. In addition, most of the biological activities of the experimental subjects are performed in vitro. Correspondingly, bioactive assays in vivo are only applied in a few studies, for example, zebrafish models used for investigations into anti-cancer/cytotoxicity [66], anti-angiogenesis [86,121], and hypoglycemia [122]. Furthermore, the difficulty of the biological screening model was another factor affecting the screening result. In fact, viruses were not considered as screening targets in general laboratory due to inherent complexity of cell-based assays of viruses [125], while mice models were expensive and time consuming. Therefore, more new natural products from the marine-derived Penicillium fungi should be screened on a wider variety of bioassays, as effective enrichment of trace compounds and enhanced methods in bioactivity screening technologies are important.

4. Conclusions

This article provided a comprehensive overview of the source, chemistry, and bioactivities of 452 secondary metabolites from marine-derived Penicillium fungi described from 2021 to 2023. Although the coronavirus disease 2019 (COVID-19) pandemic limited opportunities for field collections in domestic and international travel, the numbers of new compounds from marine-derived Penicillium fungi increased abundantly, compared to 578 new compounds reported from 1991 to 2020 [6,7]. This trend might be associated with fungal large-scale cultures under laboratory conditions and the significant impact of penicillin, the first broad-spectrum antibiotic in drug development [126]. In addition, fungal culture methods, extraction and separation techniques, structure identification technology, and biological screening methods have reached a relatively mature level [125].
New methods and in-depth research on important compounds have been carried out. Affected by the COVID-19 pandemic, both pathways of TNF-α-induced NFκB activation and TGF-β-induced Smad activation were applied to evaluate azaphilone compounds for the first time [23]. The HPLC-MS/MS analyses [9], one-pot/two-stage precursor-directed biosynthesis approach [88], and molecular networks of MS/MS data generated with Global Natural Products Social Molecular Networking (GNPS) [13] have expanded the scope of research on metabolites, especially trace components of marine-derived Penicillium fungi. Co-culture [66,97,100,107] and OSMAC [17,18,20,111] have been used to explore the structural diversity of secondary metabolites from the fungi. Further research on the known compounds, whether penicopeptide A as a candidate compound for osteoporosis prevention [12] or the anti-pancreatic cancer activity of dicitrinone G evaluated using a mouse model [11], provides an opportunity to diversify the targets, increasing the value of natural products from marine-derived Penicillium fungi.
In summary, marine-derived Penicillium fungi resources are found worldwide and have attracted great attention due to their diverse chemical structures. Penicillium fungi have produced a large number of structurally novel and bioactively potent compounds, such as polyketides, alkaloids and terpenoids. Over a thousand secondary metabolites from marine-derived Penicillium fungi have already been reported in the past thirty-three years (1991–2023). Although none of them have reached the market yet, which could partly be related to non-comprehensive screening approaches and a lack of sustained lead optimization, the mass production of trace amounts of compounds by symbiotic Penicillium fungi and the symbiotic relationship with the marine host make marine-derived Penicillium fungi a very important source of bioactive compounds for drug discovery.

Author Contributions

F.L. and Y.Z. engaged in the conceptualization and design of the manuscript. F.L. collected and analyzed the data, generated the graphs, and wrote the paper. F.L. and Y.Z. designed and drew the chemical structures. Y.Z. gave suggestions and helped in revising the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the Natural Science Foundation of Hainan (322MS131), National Natural Science Foundation of China (41776093), and Financial Fund of the Ministry of Agriculture and Rural Affairs, China (NFZX2021).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Noman, E.; Al-Shaibani, M.M.; Bakhrebah, M.A.; Almoheer, R.; Al-Sahari, M.; Al-Gheethi, A.; Mohamed, R.M.S.R.; Almulaiky, Y.Q.; Abdulaal, W.H. Potential of anti-cancer activity of secondary metabolic products from marine fungi. J. Fungi 2021, 7, 436. [Google Scholar] [CrossRef]
  2. Shin, H.J. Natural products from marine fungi. Mar. Drugs 2020, 18, 230. [Google Scholar] [CrossRef]
  3. Zhang, X.Q.; Yin, Q.Z.; Li, X.Y.; Liu, X.W.; Lei, H.X.; Wu, B. Structures and bioactivities of secondary metabolites from Penicillium genus since 2010. Fitoterapia 2022, 163, 105349. [Google Scholar] [CrossRef]
  4. Liu, S.; Su, M.Z.; Song, S.J.; Jung, J.H. Marine-derived Penicillium species as producers of cytotoxic metabolites. Mar. Drugs 2017, 15, 329. [Google Scholar] [CrossRef]
  5. Pang, K.L.; Overy, D.P.; Jones, E.B.G.; Calado, M.D.L.; Burgaud, G.; Walker, A.K.; Johnson, J.A.; Kerr, R.G.; Cha, H.J.; Bills, G.F. ‘Marine fungi’ and ‘marine-derived fungi’ in natural product chemistry research: Toward a new consensual definition. Fungal Biol. Rev. 2016, 30, 163–175. [Google Scholar] [CrossRef]
  6. Ma, H.G.; Liu, Q.; Zhu, G.L.; Liu, H.S.; Zhu, W.M. Marine natural products sourced from marine derived Penicillium fungi. J. Asian Nat. Prod. Res. 2016, 18, 92–115. [Google Scholar] [CrossRef]
  7. Yang, X.L.; Liu, J.P.; Mei, J.H.; Jiang, R.; Tu, S.Z.; Deng, H.F.; Liu, J.; Yang, S.M.; Li, J. Origins, structures, and bioactivities of secondary metabolites from marine-derived Penicillium Fungi. Mini-Rev. Med. Chem. 2021, 21, 2000–2019. [Google Scholar] [CrossRef]
  8. Zhang, P.; Wei, Q.; Yuan, X.L.; Xu, K. Newly reported alkaloids produced by marine-derived Penicillium species (covering 2014–2018). Bioorg. Chem. 2020, 99, 103840. [Google Scholar] [CrossRef]
  9. Hoang, T.P.T.; Roullier, C.; Evanno, L.; Kerzaon, I.; Gentil, E.; Pont, T.R.D.; Nazih, E.H.; Pouchus, Y.F.; Bertrand, S.; Poupon, E.; et al. Nature-inspired chemistry of complex alkaloids: Combining targeted molecular networking approach and semisynthetic strategy to access rare communesins in a marine-derived Penicillium expansum. Chem. Eur. J. 2023, 29, e202300103. [Google Scholar] [CrossRef]
  10. Fan, H.; Shi, Z.M.; Lei, Y.H.; Si-Tu, M.X.; Zhou, F.G.; Feng, C.; Wei, X.; Shao, X.H.; Chen, Y.; Zhang, C.X. Rare carbon-bridged citrinin dimers from the starfish-derived symbiotic fungus Penicillium sp. GGF16-1-2. Mar. Drugs 2022, 20, 443. [Google Scholar] [CrossRef]
  11. Shi, Z.M.; Zhang, M.Y.; Fan, H.; Chen, Y.J.; Dong, S.; Zhou, F.G.; Wang, B.; Liu, J.Y.; Jin, J.Q.; Luo, Y.; et al. The marine Penicillium sp. GGF16-1-2 metabolite dicitrinone G inhibits pancreatic angiogenesis by regulating the activation of NLRP3 inflammasome. J. Nat. Med. 2024, 78, 78–90. [Google Scholar] [CrossRef] [PubMed]
  12. Xie, C.L.; Yue, Y.T.; Xu, J.P.; Li, N.; Lin, T.; Ji, G.R.; Yang, X.W.; Xu, R. Penicopeptide A (PPA) from the deep-sea-derived fungus promotes osteoblast-mediated bone formation and alleviates ovariectomy-induced bone loss by activating the AKT/GSK-3β/β-catenin signaling pathway. Pharmacol. Res. 2023, 197, 106968. [Google Scholar] [CrossRef]
  13. Hao, B.C.; Zheng, Y.Y.; Li, Z.H.; Zheng, C.J.; Wang, C.Y.; Chen, M. Targeted isolation of prenylated indole alkaloids from the marine-derived fungus Penicillium janthinellum HK1-6 using molecular networking. Nat. Prod. Res. 2023. [Google Scholar] [CrossRef]
  14. Chen, T.; Yang, W.C.; Li, T.B.; Yin, Y.H.; Liu, Y.F.; Wang, B.; She, Z.G. Hemiacetalmeroterpenoids A–C and astellolide Q with antimicrobial activity from the marine-derived fungus Penicillium sp. N-5. Mar. Drugs 2022, 20, 514. [Google Scholar] [CrossRef]
  15. Wang, Y.; Chen, W.H.; Xu, Z.F.; Bai, Q.Q.; Zhou, X.M.; Zheng, C.J.; Bai, M.; Chen, G.Y. Biological secondary metabolites from the Lumnitzera littorea-derived fungus Penicillium oxalicum HLLG-13. Mar. Drugs 2023, 21, 22. [Google Scholar] [CrossRef]
  16. Bai, M.; Gao, C.H.; Liu, K.; Zhao, L.Y.; Tang, Z.Z.; Liu, Y.H. Two new benzophenones isolated from a mangrove-derived fungus Penicillium sp. J. Antibiot. 2021, 74, 821–824. [Google Scholar] [CrossRef]
  17. Zeng, Y.B.; Wang, Z.; Chang, W.J.; Zhao, W.B.; Wang, H.; Chen, H.Q.; Dai, H.F.; Lv, F. New azaphilones from the marine-derived fungus Penicillium sclerotiorum E23Y-1A with their anti-inflammatory and antitumor activities. Mar. Drugs 2023, 21, 75. [Google Scholar] [CrossRef]
  18. Wang, Z.; Zeng, Y.B.; Zhao, W.B.; Dai, H.F.; Chang, W.J.; Lv, F. Structures and biological activities of brominated azaphilones produced by Penicillium sclerotiorum E23Y-1A. Phytochem. Lett. 2022, 52, 138–142. [Google Scholar] [CrossRef]
  19. Hu, X.Y.; Li, X.M.; Wang, B.G.; Meng, L.H. Uncommon polyketides from Penicillium steckii AS-324, a marine endozoic fungus isolated from deep-sea coral in the magellan seamount. Int. J. Mol. Sci. 2022, 23, 6332. [Google Scholar] [CrossRef]
  20. Chen, S.R.; Wang, S.W.; Chen, C.Y.; Ke, T.Y.; Lin, J.J.; Hwang, T.L.; Huang, Y.T.; Huang, Y.C.; Cheng, Y.B. Additional azaphilones from the marine algae-derived fungus Penicillium sclerotiorum with anti-angiogenic activity. Bull. Chem. Soc. Jpn. 2023, 96, 1–7. [Google Scholar] [CrossRef]
  21. Yao, F.H.; Liang, X.; Qi, S.H. Two new linear peptides from the marine-derived fungus Penicillium sp. SCSIO 41512. J. Asian Nat. Prod. Res. 2023, 25, 941–958. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, S.; Zeng, Y.B.; Yin, J.J.; Chang, W.J.; Zhao, X.L.; Mao, Y. Two new azaphilones from the marine-derived fungus Penicillium sclerotiorum E23Y-1A. Phytochem. Lett. 2022, 47, 76–80. [Google Scholar] [CrossRef]
  23. Wang, H.C.; Ke, T.Y.; Ko, Y.C.; Lin, J.J.; Chang, J.S.; Cheng, Y.B. Anti-inflammatory azaphilones from the edible alga-derived fungus Penicillium sclerotiorum. Mar. Drugs 2021, 19, 529. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, W.; Wang, M.; Wang, X.B.; Li, Y.Q.; Ding, J.L.; Lan, M.X.; Gao, X.; Zhao, D.L.; Zhang, C.S.; Wu, G.X. Phytotoxic azaphilones from the mangrove-derived fungus Penicillium sclerotiorum HY5. Front. Microbiol. 2022, 13, 880874. [Google Scholar] [CrossRef] [PubMed]
  25. Zhang, J.; Chen, Y.C.; Liu, Z.M.; Li, S.N.; Zhong, J.Q.; Guo, B.H.; Liu, H.X.; Zhang, W.M. Azaphilones and isocoumarin derivatives from Penicillum chermesinum FS625 isolated from the South China Sea. Tetrahedron Lett. 2021, 73, 153117. [Google Scholar] [CrossRef]
  26. Zhang, H.; Lei, X.X.; Shao, S.R.; Zhou, X.F.; Li, Y.Q.; Yang, B. Azaphilones and meroterpenoids from the soft coral-derived fungus Penicillium glabrum glmu003. Chem. Biodivers. 2021, 18, e2100663. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, C.; Ye, G.T.; Tang, J.; Li, J.L.; Liu, W.B.; Wu, L.; Long, Y.H. New polyketides from mangrove endophytic fungus Penicillium sp. BJR-P2 and their anti-inflammatory activity. Mar. Drugs 2022, 20, 583. [Google Scholar] [CrossRef] [PubMed]
  28. Gan, Y.M.; Xia, J.L.; Zhao, L.Y.; Liu, K.; Tang, Z.Z.; Huang, B.Y.; Liu, Y.H.; Gao, C.H.; Bai, M. Two new isocoumarins isolated from a mangrove-derived Penicillium sp. Phytochem. Lett. 2022, 50, 21–24. [Google Scholar] [CrossRef]
  29. Chu, Y.C.; Chang, C.H.; Liao, H.R.; Cheng, M.J.; Wu, M.D.; Fu, S.L.; Chen, J.J. Rare chromone derivatives from the marine-derived Penicillium citrinum with anti-cancer and anti-inflammatory activities. Mar. Drugs 2021, 19, 25. [Google Scholar] [CrossRef]
  30. Chu, Y.C.; Chang, C.H.; Liao, H.R.; Fu, S.L.; Chen, J.J. Anti-cancer and anti-inflammatory activities of three new chromone derivatives from the marine-derived Penicillium citrinum. Mar. Drugs 2021, 19, 408. [Google Scholar] [CrossRef]
  31. Han, S.Y.; Liu, Y.; Liu, W.; Yang, F.; Zhang, J.; Liu, R.F.; Zhao, F.Q.; Xu, W.; Cheng, Z.B. Chromone derivatives with α-glucosidase inhibitory activity from the marine fungus Penicillium thomii Maire. Molecules 2021, 26, 5273. [Google Scholar] [CrossRef] [PubMed]
  32. Yang, F.; Liu, Y.; Zhang, X.Q.; Liu, W.; Qiao, Y.; Xu, W.; Li, Q.; Cheng, Z.B. Three new chromone derivatives from the deep-sea-derived fungus Penicillium thomii. Rec. Nat. Prod. 2023, 17, 174–178. [Google Scholar]
  33. Lai, C.R.; Tian, D.M.; Zheng, M.X.; Li, B.L.; Jia, J.; Wei, J.H.; Wu, B.; Bi, H.K.; Tang, J.S. Novel citrinin derivatives from fungus Penicillium sp. TW131-64 and their antimicrobial activities. Appl. Microbiol. Biot. 2023, 107, 6607–6619. [Google Scholar] [CrossRef] [PubMed]
  34. He, Z.H.; Xie, C.L.; Wu, T.Z.; Zhang, Y.; Zou, Z.B.; Xie, M.M.; Xu, L.; Capon, R.J.; Xu, R.; Yang, X.W. Neotricitrinols A-C, unprecedented citrinin trimers with anti-osteoporosis activity from the deep-sea-derived Penicillium citrinum W23. Bioorg. Chem. 2023, 139, 106756. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, S.J.; Tian, D.M.; Wei, J.H.; Li, C.; Ma, Y.H.; Gou, X.S.; Shen, Y.R.; Chen, M.; Zhang, S.H.; Li, J.; et al. Citrinin derivatives from Penicillium citrinum Y34 that inhibit α-glucosidase and ATP-citrate lyase. Front. Mar. Sci. 2022, 9, 961356. [Google Scholar] [CrossRef]
  36. Han, W.R.; Song, M.M.; Hu, Y.W.; Pang, X.Y.; Liao, S.R.; Yang, B.; Zhou, X.F.; Liu, Y.H.; Liu, Q.C.; Wang, J.F. Citrinin and α-pyrone derivatives with pancreatic lipase inhibitory activities from Penicillium sp. SCSIO 41302. J. Asian Nat. Prod. Res. 2022, 24, 810–819. [Google Scholar] [CrossRef] [PubMed]
  37. Guo, T.T.; Song, M.M.; Han, W.R.; Zhu, J.H.; Liu, Q.C.; Wang, J.F. New N-methyl-4-quinolone alkaloid and citrinin dimer derivatives from the sponge-derived fungus Penicillium sp. SCSIO 41303. Phytochem. Lett. 2021, 46, 29–35. [Google Scholar] [CrossRef]
  38. Leshchenko, E.V.; Antonov, A.S.; Borkunov, G.V.; Hauschild, J.; Zhuravleva, O.I.; Khudyakova, Y.V.; Menshov, A.S.; Popov, R.S.; Kim, N.Y.; Graefen, M.; et al. New bioactive β-resorcylic acid derivatives from the alga-derived fungus Penicillium antarcticum KMM 4685. Mar. Drugs 2023, 21, 178. [Google Scholar] [CrossRef] [PubMed]
  39. Li, Y.H.; Li, X.M.; Li, X.; Yang, S.Q.; Wang, B.G.; Li, H.L. Verrucosidin derivatives from the deep sea cold-seep-derived fungus Penicillium polonicum CS-252. Int. J. Mol. Sci. 2022, 23, 5567. [Google Scholar] [CrossRef]
  40. Li, Y.H.; Mándi, A.; Li, H.L.; Li, X.M.; Li, X.; Meng, L.H.; Yang, S.Q.; Shi, X.S.; Kurtán, T.; Wang, B.G. Isolation and characterization of three pairs of verrucosidin epimers from the marine sediment-derived fungus Penicillium cyclopium and configuration revision of penicyrone A and related analogues. Mar. Life Sci. Technol. 2023, 5, 223–231. [Google Scholar] [CrossRef]
  41. Li, Y.H.; Yang, S.Q.; Li, X.M.; Li, X.; Wang, B.G.; Li, H.L. Five new verrucosidin derivatives from Penicillium polonicum, a deep-sea cold-seep sediment isolated fungus. Fitoterapia 2023, 165, 105387. [Google Scholar] [CrossRef]
  42. Xing, C.P.; Chen, D.; Xie, C.L.; Liu, Q.M.; Zhong, T.H.; Shao, Z.Z.; Liu, G.M.; Luo, L.Z.; Yang, X.W. Anti-food allergic compounds from Penicillium griseofulvum MCCC 3A00225, a deep-sea-derived fungus. Mar. Drugs 2021, 19, 224. [Google Scholar] [CrossRef]
  43. Zou, Z.B.; Zhang, G.; Zhou, Y.Q.; Xie, C.L.; Xie, M.M.; Xu, L.; Hao, Y.J.; Luo, L.Z.; Zhang, X.K.; Yang, X.W.; et al. Chemical constituents of the deep-sea-derived Penicillium citreonigrum MCCC 3A00169 and their antiproliferative effects. Mar. Drugs 2022, 20, 736. [Google Scholar] [CrossRef]
  44. Li, H.C.; Zhang, W.; Zhang, X.; Tang, S.; Men, P.; Xiong, M.Y.; Li, Z.M.; Zhang, Y.Y.; Huang, X.N.; Lu, X.F. Identification of PKS-NRPS hybrid metabolites in marine-derived Penicillium oxalicum. Mar. Drugs 2022, 20, 523. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, S.Z.; He, F.M.; Bin, Y.L.; Li, C.F.; Xie, B.Y.; Tang, X.X.; Qiu, Y.K. Bioactive compounds derived from the marine-derived fungus MCCC3A00951 and their influenza neuraminidase inhibition activity in vitro and in silico. Nat. Prod. Res. 2021, 35, 5621–5628. [Google Scholar] [CrossRef] [PubMed]
  46. Song, Y.Y.; She, J.L.; Chen, W.H.; Wang, J.M.; Tan, Y.H.; Pang, X.Y.; Zhou, X.F.; Wang, J.F.; Liu, Y.H. New fusarin derivatives from the marine algicolous fungus Penicillium steckii SCSIO41040. Mar. Drugs 2023, 21, 532. [Google Scholar] [CrossRef]
  47. Pang, X.Y.; Wang, P.; Liao, S.R.; Zhou, X.F.; Lin, X.P.; Yang, B.; Tian, X.P.; Wang, J.F.; Liu, Y.H. Three unusual hybrid sorbicillinoids with anti-inflammatory activities from the deep-sea derived fungus Penicillium sp. SCSIO06868. Phytochemistry 2022, 202, 113311. [Google Scholar] [CrossRef]
  48. Pang, X.Y.; Zhou, X.F.; Lin, X.P.; Yang, B.; Tian, X.P.; Wang, J.F.; Xu, S.H.; Liu, Y.H. Structurally various sorbicillinoids from the deep-sea sediment derived fungus Penicillium sp. SCSIO06871. Bioorg. Chem. 2021, 107, 104600. [Google Scholar] [CrossRef]
  49. Xie, C.L.; Zhang, D.; Lin, T.; He, Z.H.; Yan, Q.X.; Cai, Q.; Zhang, X.K.; Yang, X.W.; Chen, H.F. Antiproliferative sorbicillinoids from the deep-sea-derived Penicillium allii-sativi. Front. Microbiol. 2021, 11, 636948. [Google Scholar] [CrossRef] [PubMed]
  50. Ding, W.J.; Wang, F.F.; Li, Q.W.; Xue, Y.X.; Dong, Z.T.; Tian, D.M.; Chen, M.; Zhang, Y.W.; Hong, K.; Tang, J.S. Isolation and characterization of anti-inflammatory sorbicillinoids from the mangrove-derived fungus Penicillium sp. DM815. Chem. Biodivers. 2021, 18, e2100229. [Google Scholar] [CrossRef] [PubMed]
  51. Wu, X.Z.; Huang, W.J.; Liu, W.; Mándi, A.; Zhang, Q.B.; Zhang, L.P.; Zhang, W.J.; Kurtán, T.; Yuan, C.S.; Zhang, C.S. Penicisteckins A-F, isochroman-derived atropisomeric dimers from Penicillium steckii HNNU-5B18. J. Nat. Prod. 2021, 84, 2953–2960. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, L.; Shi, Y.Q.; Che, Q.; Zhu, T.J.; Zhang, G.J.; Zhang, X.K.; Li, M.Y.; Li, D.H. Penipyrols C-G and methyl-penipyrol A, α-pyrone polyketides from the mangrove derived fungus Penicillium sp. HDN-11-131. Bioorg. Chem. 2021, 113, 104975. [Google Scholar] [CrossRef] [PubMed]
  53. Ding, Y.; Jiang, Y.; Xu, S.J.; Xin, X.J.; An, F.L. Perpyrrospirone A, an unprecedented hirsutellone peroxide from the marine-derived Penicillium citrinum. Chin. Chem. Lett. 2023, 34, 107562. [Google Scholar] [CrossRef]
  54. Hao, Y.J.; Zou, Z.B.; Xie, M.M.; Zhang, Y.; Xu, L.; Yu, H.Y.; Ma, H.B.; Yang, X.W. Ferroptosis inhibitory compounds from fungus Penicillium sp. MCCC 3A00126. Mar. Drugs 2023, 21, 234. [Google Scholar] [CrossRef] [PubMed]
  55. Cao, G.P.; Xia, J.L.; Zhao, L.Y.; Tang, Z.Z.; Lin, X.; Liu, Y.H.; Gao, C.H.; Liu, K.; Bai, M. Penicixanthene E, a new xanthene isolated from a mangrove-derived fungus Penicillium sp. J. Antibiot. 2022, 75, 526–529. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, Y.F.; Lu, Y.; Zhao, J.Y.; Chen, Q. Polyketides and alkaloids from the fungus Penicillium sp. Rec. Nat. Prod. 2023, 17, 367–371. [Google Scholar] [CrossRef]
  57. Zhang, Y.; Xie, C.L.; Wang, Y.; He, X.W.; Xie, M.M.; Li, Y.; Zhang, K.; Zou, Z.B.; Yang, L.H.; Xu, R.; et al. Penidihydrocitrinins A-C: New polyketides from the deep-sea-derived Penicillium citrinum W17 and their anti-inflammatory and anti-osteoporotic bioactivities. Mar. Drugs 2023, 21, 538. [Google Scholar] [CrossRef] [PubMed]
  58. Qi, X.Y.; Liu, B.; Jiang, Z.X. A new cytotoxic phenalenone derivative from Penicillium oxalicum. Nat. Prod. Res. 2023, 37, 1397–1400. [Google Scholar] [CrossRef] [PubMed]
  59. Huo, R.Y.; Zhang, J.X.; Niu, S.B.; Liu, L. New prenylated indole diketopiperazine alkaloids and polyketides from the mangrovederived fungus Penicillium sp. Front. Mar. Sci. 2022, 9, 1097594. [Google Scholar] [CrossRef]
  60. Gou, X.S.; Tian, D.M.; Wei, J.H.; Ma, Y.H.; Zhang, Y.X.; Chen, M.; Ding, W.J.; Wu, B.; Tang, J.S. New drimane sesquiterpenes and polyketides from marine-derived fungus Penicillium sp. TW58-16 and their anti-inflammatory and α-glucosidase inhibitory effects. Mar. Drugs 2021, 19, 416. [Google Scholar] [CrossRef]
  61. Pang, X.Y.; Chen, W.H.; Wang, X.; Zhou, X.F.; Yang, B.; Tian, X.P.; Wang, J.F.; Xu, S.H.; Liu, Y.H. New tetramic acid derivatives from the deep-sea-derived fungus Penicillium sp. SCSIO06868 with SARS-CoV-2 Mpro inhibitory activity evaluation. Front. Microbiol. 2021, 12, 730807. [Google Scholar] [CrossRef] [PubMed]
  62. Tiana, D.M.; Goua, X.S.; Jia, J.; Wei, J.H.; Zheng, M.X.; Ding, W.J.; Bib, H.K.; Wu, B.; Tang, J.S. New diketopiperazine alkaloid and polyketides from marine-derived fungus Penicillium sp. TW58-16 with antibacterial activity against Helicobacter pylori. Fitoterapia 2022, 156, 105095. [Google Scholar] [CrossRef] [PubMed]
  63. Song, Y.Y.; Tan, Y.H.; She, J.L.; Chen, C.M.; Wang, J.M.; Hu, Y.W.; Pang, X.Y.; Wang, J.F.; Liu, Y.H. Tanzawaic acid derivatives from the marine-derived Penicillium steckii as inhibitors of RANKL-induced osteoclastogenesis. J. Nat. Prod. 2023, 86, 1171–1178. [Google Scholar] [CrossRef] [PubMed]
  64. Hu, X.Y.; Li, X.M.; Wang, B.G.; Meng, L.H. Tanzawaic acid derivatives: Fungal polyketides from the deep-Sea coral-derived endozoic Penicillium steckii AS-324. J. Nat. Prod. 2022, 85, 1398–1406. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, C.M.; Chen, W.H.; Tao, H.M.; Yang, B.; Zhou, X.F.; Luo, X.W.; Liu, Y.H. Diversified polyketides and nitrogenous compounds from the mangrove endophytic fungus Penicillium steckii SCSIO 41025. Chin. J. Chem. 2021, 39, 2132–2140. [Google Scholar] [CrossRef]
  66. Yu, G.H.; Sun, P.; Aierken, R.; Sun, C.X.; Zhang, Z.Z.; Che, Q.; Zhang, G.J.; Zhu, T.J.; Gu, Q.Q.; Li, M.Y.; et al. Linear polyketides produced by co-culture of Penicillium crustosum and Penicillium fellutanum. Mar. Life Sci. Technol. 2021, 4, 237–244. [Google Scholar] [CrossRef]
  67. Ying, Z.; Li, X.M.; Yang, S.Q.; Wang, B.G.; Li, H.L.; Meng, L.H. New polyketide and sesquiterpenoid derivatives from the magellan seamount-derived fungus Penicillium rubens AS-130. Chem. Biodivers. 2023, 20, e202300229. [Google Scholar] [CrossRef] [PubMed]
  68. Weng, W.Y.; Li, R.D.; Zhang, Y.X.; Pan, X.F.; Jiang, S.C.; Sun, C.C.; Zhang, C.; Lu, X.M. Polyketides isolated from an endophyte Penicillium oxalicum 2021CDF-3 inhibit pancreatic tumor growth. Front. Microbiol. 2022, 13, 1033823. [Google Scholar] [CrossRef] [PubMed]
  69. He, Z.H.; Wu, J.; Xu, L.; Hu, M.Y.; Xie, M.M.; Hao, Y.J.; Li, S.J.; Shao, Z.Z.; Yang, X.W. Chemical constituents of the deep-sea-derived Penicillium solitum. Mar. Drugs 2021, 19, 580. [Google Scholar] [CrossRef]
  70. Dyshlovoy, S.A.; Zhuravleva, O.I.; Hauschild, J.; Busenbender, T.; Pelageev, D.N.; Yurchenko, A.N.; Khudyakova, Y.V.; Antonov, A.S.; Graefen, M.; Bokemeyer, C.; et al. New Marine fungal deoxy-14,15-dehydroisoaustamide resensitizes prostate cancer cells to enzalutamide. Mar. Drugs 2023, 21, 54. [Google Scholar] [CrossRef]
  71. Zhuravleva, O.I.; Antonov, A.S.; Trang, V.T.D.; Pivkin, M.V.; Khudyakova, Y.V.; Denisenko, V.A.; Popov, R.S.; Kim, N.Y.; Yurchenko, E.A.; Gerasimenko, A.V.; et al. New deoxyisoaustamide derivatives from the coral-derived fungus Penicillium dimorphosporum KMM 4689. Mar. Drugs 2021, 19, 32. [Google Scholar] [CrossRef] [PubMed]
  72. Hu, Y.W.; Chen, W.H.; Song, M.M.; Pang, X.Y.; Tian, X.P.; Wang, F.Z.; Liu, Y.H.; Wang, J.F. Indole diketopiperazine alkaloids and aromatic polyketides from the Antarctic fungus Penicillium sp. SCSIO 05705. Nat. Prod. Res. 2023, 37, 389–396. [Google Scholar] [CrossRef]
  73. Zhang, J.X.; Zhang, B.D.; Shi, Y.; Zhai, Y.N.; Ren, J.W.; Cai, L.; Sun, L.Y.; Liu, L. Penindolacid A, a new indole alkaloid from the marine derived fungus Penicillium sp. Magn. Reson. Chem. 2023, 61, 554–559. [Google Scholar] [CrossRef]
  74. Yong, K.; Kaleem, S.; Ma, M.Z.; Lian, X.Y.; Zhang, Z.Z. Antiglioma natural products from the marine-associated fungus Penicillium sp. ZZ1750. Molecules 2022, 27, 7099. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, C.Z.; Li, G.; Zhang, Y.H.; Yuan, S.Z.; Dong, K.M.; Lou, H.X.; Peng, X.P. Interconvertible pyridone alkaloids from the marine-derived fungus Penicillium oxalicum QDU1. J. Nat. Prod. 2023, 86, 739–750. [Google Scholar] [CrossRef] [PubMed]
  76. Xu, F.Q.; Chen, W.H.; Ye, Y.X.; Qi, X.; Zhao, K.; Long, J.Y.; Pang, X.Y.; Liu, Y.H.; Wang, J.F. A new quinolone and acetylcholinesterase inhibitors from a sponge-associated fungus Penicillium sp. SCSIO41033. Nat. Prod. Res. 2023, 37, 2871–2877. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, Y.Y.; Xue, X.Y.; Zhou, L.J.; Yang, W.C.; She, Z.G.; Liao, Q.N.; Feng, Y.K.; Chen, X.K.; Zhang, Y. Quinolinones alkaloids with AChE inhibitory activity from mangrove endophytic fungus Penicillium citrinum YX-002. Chem. Biodivers. 2023, 20, e202300735. [Google Scholar] [CrossRef]
  78. Chen, C.M.; Chen, W.H.; Pang, X.Y.; Liao, S.R.; Wang, J.F.; Lin, X.P.; Yang, B.; Zhou, X.F.; Luo, X.W.; Liu, Y.H. Pyrrolyl 4-quinolone alkaloids from the mangrove endophytic fungus Penicillium steckii SCSIO 41025: Chiral resolution, configurational assignment, and enzyme inhibitory activities. Phytochemistry 2021, 186, 112730. [Google Scholar] [CrossRef] [PubMed]
  79. Yao, F.H.; Liang, X.; Lu, X.H.; Cheng, X.; Luo, L.X.; Qi, S.H. Pyrrospirones K-Q, decahydrofluorene-class alkaloids from the marine-derived fungus Penicillium sp. SCSIO 41512. J. Nat. Prod. 2022, 85, 2071–2208. [Google Scholar] [CrossRef]
  80. Cai, J.; Wang, X.N.; Yang, Z.Z.; Tan, Y.H.; Peng, B.; Liu, Y.H.; Zhou, X.F. Thiodiketopiperazines and alkane derivatives produced by the mangrove sediment-derived fungus Penicillium ludwigii SCSIO 41408. Front. Microbiol. 2022, 13, 857041. [Google Scholar] [CrossRef]
  81. Jiang, G.D.; Zhang, P.L.; Ratnayake, R.; Yang, G.; Zhang, Y.; Zuo, R.; Powell, M.; Huguet-Tapia, J.C.; Abboud, K.A.; Dang, L.H.; et al. Fungal epithiodiketopiperazines carrying α, β-polysulfide bridges from Penicillium steckii YE, and their chemical interconversion. ChemBioChem 2021, 21, 416–422. [Google Scholar] [CrossRef] [PubMed]
  82. Ye, Y.X.; Liang, J.Q.; She, J.L.; Lin, X.P.; Wang, J.F.; Liu, Y.H.; Yang, D.H.; Tan, Y.H.; Luo, X.W.; Zhou, X.F. Two new alkaloids and a new butenolide derivative from the beibu gulf sponge-derived fungus Penicillium sp. SCSIO 41413. Mar. Drugs 2023, 21, 27. [Google Scholar] [CrossRef] [PubMed]
  83. Li, H.M.; Long, J.Y.; Wang, X.N.; She, J.L.; Liu, Y.H.; Li, Y.Q.; Yang, B. Bioactive secondary metabolites isolated from the soft coral derived Penicillium sp. SCSIO 41038. Nat. Prod. Res. 2023. [Google Scholar] [CrossRef] [PubMed]
  84. Yu, H.Y.; Li, Y.; Zhang, M.; Zou, Z.B.; Hao, Y.J.; Xie, M.M.; Li, L.S.; Meng, D.L.; Yang, X.W. Chemical Constituents of the deep-sea gammarid shrimp-derived fungus Penicillium citrinum XIA-16. Chem. Biodivers. 2023, 20, e202301507. [Google Scholar] [CrossRef]
  85. Jiang, J.Y.; Jiang, H.M.; Shen, D.N.; Chen, Y.C.; Shi, H.J.; He, F. Citrinadin C, a new cytotoxic pentacyclic alkaloid from marine-derived fungus Penicillium citrinum. J. Antibiot. 2022, 75, 301–303. [Google Scholar] [CrossRef] [PubMed]
  86. Qiu, Y.Z.; Zhu, Y.Q.; Lu, H.; Li, X.B.; Liu, K.C.; Li, P.H.; Wang, L.Z.; Zhang, X.M.; Chen, H.; Lin, H.W.; et al. Secondary metabolites from the marine-derived fungus Penicillium chrysogenum Y20-2, and their pro-angiogenic activity. Z. Naturforschung C 2023, 78, 345–352. [Google Scholar] [CrossRef]
  87. Chen, J.; Huo, L.N.; Gao, Y.; Zhang, Y.L.; Che, Y. Two new N-acetyl-D-glucosamine derivatives from the medical algae-derived endophytic fungus Penicillium chrysogenum. Nat. Prod. Res. 2022, 36, 3988–3991. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, Z.Z.; He, X.Q.; Zhang, X.M.; Li, D.H.; Wu, G.W.; Liu, Z.Z.; Niu, C.; Yang, L.P.; Song, W.T.; Li, Z.L.; et al. Production of multiple talaroenamines from Penicillium malacosphaerulum via one-pot/two-stage precursor-directed biosynthesis. J. Nat. Prod. 2022, 85, 2168–2176. [Google Scholar] [CrossRef]
  89. Li, S.H.; Ma, Y.H.; Wang, L.X.; Lan, D.H.; Fu, L.L.; Wu, B. Two new alkaloids from the marine-derived fungus Penicillium sp. LSH-3-1. Chem. Biodivers. 2022, 19, e202200310. [Google Scholar] [CrossRef] [PubMed]
  90. Chen, C.Y.; Qi, J.F.; He, Y.J.; Lu, Y.Y.; Wang, Y. Genomic and chemical profiling of B9, a unique Penicillium fungus derived from sponge. J. Fungi 2022, 8, 686. [Google Scholar] [CrossRef] [PubMed]
  91. Yang, J.F.; Song, Y.X.; Zhou, Z.B.; Huang, Y.; Wang, S.T.; Yuan, J.; Wong, N.K.; Yan, Y.; Ju, J.H. Sulfoxanthicillin from the deep-sea derived Penicillium sp. SCSIO sof101: An antimicrobial compound against gram-positive and -negative pathogens. J. Antibiot. 2023, 76, 113–120. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, J.P.; Liu, D.; Fan, A.L.; Huang, J.; Lin, W.H. Eremophilane-type sesquiterpenes from a marine-derived fungus Penicillium copticola with antitumor and neuroprotective activities. Mar. Drugs 2022, 20, 712. [Google Scholar] [CrossRef] [PubMed]
  93. Huang, Q.R.; Wang, Y.H.; Chi, X.Y.; Liu, C.; Zhang, J.L. A new drimane sesquiterpene ester from the marine-derived fungus Penicillium chrysogenum LD-201810. Chem. Nat. Compd. 2022, 58, 1042–1044. [Google Scholar] [CrossRef]
  94. Kaleem, S.; Ge, H.J.; Yi, W.W.; Zhang, Z.Z.; Wu, B. Isolation, structural elucidation, and antimicrobial evaluation of the metabolites from a marine-derived fungus Penicillium sp. ZZ1283. Nat. Prod. Res. 2021, 35, 2498–2506. [Google Scholar] [CrossRef] [PubMed]
  95. Zhang, W.F.; Meng, Q.Y.; Wu, J.S.; Cheng, W.; Liu, D.; Huang, J.; Fan, A.L.; Xu, J.; Lin, W.H. Acorane sesquiterpenes from the deep-sea derived Penicillium bilaiae fungus with anti-neuroinflammatory effects. Front. Chem. 2022, 10, 1036212. [Google Scholar] [CrossRef] [PubMed]
  96. Wu, Q.; Chang, Y.M.; Che, Q.; Li, D.H.; Zhang, G.J.; Zhu, T.J. Citreobenzofuran D-F and phomenone A-B: Five novel sesquiterpenoids from the mangrove-derived fungus Penicillium sp. HDN13-494. Mar. Drugs 2022, 20, 137. [Google Scholar] [CrossRef] [PubMed]
  97. Li, X.Y.; Ge, Y.C.; Ma, Y.H.; Wang, S.B.; Li, S.H.; Yin, Q.Z.; Liu, X.W.; Wie, J.H.; Wu, X.D.; Wu, B. New cytotoxic secondary metabolites from two deep-sea-derived fungi and the co-culture impact on the secondary metabolic patterns. Chem. Biodivers. 2022, 19, e202200055. [Google Scholar] [CrossRef] [PubMed]
  98. Ge, Y.; Tang, W.L.; Huang, Q.R.; Wei, M.L.; Li, Y.Z.; Jiang, L.L.; Li, C.L.; Yu, X.; Zhu, H.W.; Chen, G.Z.; et al. New enantiomers of a nor-bisabolane derivative and two new phthalides produced by the marine-derived fungus Penicillium chrysogenum LD-201810. Front. Microbiol. 2021, 12, 727670. [Google Scholar] [CrossRef] [PubMed]
  99. Pang, S.; Guo, Z.G.; Wang, L.; Guo, Q.F.; Cao, F. Anti-IAV indole-diterpenoids from the marine derived fungus Penicillium citrinum. Nat. Prod. Res. 2023, 37, 586–591. [Google Scholar] [CrossRef]
  100. Cao, F.; Liu, X.M.; Wang, X.; Zhang, Y.H.; Yang, J.; Li, W.; Luo, D.Q.; Liu, Y.F. Structural diversity and biological activities of indole-diterpenoids from Penicillium janthinellum by co-culture with Paecilomyces formosus. Bioorg. Chem. 2023, 141, 106863. [Google Scholar] [CrossRef]
  101. Zhang, Y.H.; Li, L.; Li, Y.Q.; Luo, J.H.; Li, W.; Li, L.F.; Zheng, C.J.; Cao, F. Oxalierpenes A and B, unusual indole-diterpenoid derivatives with antiviral activity from a marine-derived strain of the fungus Penicillium oxalicum. J. Nat. Prod. 2022, 85, 1880–1885. [Google Scholar] [CrossRef]
  102. Yurchenko, A.N.; Zhuravleva, O.I.; Khmel, O.O.; Oleynikova, G.K.; Antonov, A.S.; Kirichuk, N.N.; Chausova, V.E.; Kalinovsky, A.I.; Berdyshev, D.V.; Kim, N.Y.; et al. New cyclopiane diterpenes and polyketide derivatives from marine sediment-derived fungus Penicillium antarcticum KMM 4670 and their biological activities. Mar. Drugs 2023, 21, 584. [Google Scholar] [CrossRef]
  103. Wang, X.X.; Chen, Z.L.; Zhang, J.S.; Liu, H.S.; Ma, R.P.; Liu, X.P.; Li, M.Y.; Ge, D.; Bao, J.; Zhang, H. Indole diterpenes from mangrove sediment-derived fungus Penicillium sp. UJNMF0740 protect PC12 cells against 6-OHDA-induced neurotoxicity via regulating the PI3K/Akt pathway. Mar. Drugs 2023, 21, 593. [Google Scholar] [CrossRef] [PubMed]
  104. Dai, L.T.; Yang, L.; Kong, F.D.; Ma, Q.Y.; Xie, Q.Y.; Dai, H.F.; Yu, Z.F.; Zhao, Y.X. Cytotoxic Indole-diterpenoids from the marine-derived fungus Penicillium sp. KFD28. Mar. Drugs 2021, 19, 613. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, M.Y.; Xie, Q.Y.; Kong, F.D.; Ma, Q.Y.; Zhou, L.M.; Yuan, J.Z.; Dai, H.F.; Wu, Y.G.; Zhao, Y.X. Two new indole-diterpenoids from the marine-derived fungus Penicillium sp. KFD28. J. Asian Nat. Prod. Res. 2021, 23, 1030–1036. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, X.; Zhao, M.; Chen, J.; Pan, W.C.; Tan, S.L.; Cui, H.; Zhao, Z.X. Seven new meroterpenoids from the fungus Penicillium sclerotiorum GZU-XW03–2. Fitoterapia 2023, 165, 105428. [Google Scholar] [CrossRef] [PubMed]
  107. Eze, P.M.; Liu, Y.; Simons, V.E.; Ebada, S.S.; Kurt’an, T.; Kir’aly, S.B.; Esimone, C.O.; Okoye, F.B.C.; Proksch, P.; Kalscheuer, R. Two new metabolites from a marine-derived fungus Penicillium ochrochloron. Phytochem. Lett. 2023, 55, 101–104. [Google Scholar] [CrossRef]
  108. Hu, X.Y.; Li, X.M.; Liu, H.; Wang, B.G.; Meng, L.H. Mining new meroterpenoids from the marine red alga-derived endophytic Penicillium chermesinum EN-480 by comparative transcriptome analysis. Bioorg. Chem. 2022, 128, 106021. [Google Scholar] [CrossRef]
  109. Zhao, M.; Chen, X.C.; Pan, W.C.; Liu, X.; Tan, S.L.; Cui, H. Meroterpenoids from the fungus Penicillium sclerotiorum GZU-XW03-2 and their anti-inflammatory activity. Phytochemistry 2022, 202, 113307. [Google Scholar] [CrossRef]
  110. Xie, C.L.; Zhang, D.; Guo, K.Q.; Yan, Q.X.; Zou, Z.B.; He, Z.H.; Wu, Z.; Zhang, X.K.; Chen, H.F.; Yang, X.W. Meroterpenthiazole A, a unique meroterpenoid from the deep-sea-derived Penicillium allii-sativi, significantly inhibited retinoid X receptor (RXR)-α transcriptional effect. Chin. Chem. Lett. 2022, 33, 2057–2059. [Google Scholar] [CrossRef]
  111. Xie, C.L.; Liu, Q.M.; He, Z.H.; Gai, Y.B.; Zou, Z.B.; Shao, Z.Z.; Liu, G.M.; Chen, H.F.; Yang, X.W. Discovery of andrastones from the deep-sea-derived Penicillium allii-sativi MCCC 3A00580 by OSMAC strategy. Bioorg. Chem. 2021, 108, 104671. [Google Scholar] [CrossRef] [PubMed]
  112. Ren, J.W.; Huo, R.Y.; Liu, G.R.; Liu, L. New andrastin-type meroterpenoids from the marine-derived fungus Penicillium sp. Mar. Drugs 2021, 19, 189. [Google Scholar] [CrossRef] [PubMed]
  113. Bai, M.; Zheng, C.J.; Chen, G.Y. Austins-type meroterpenoids from a mangrove-derived Penicillium sp. J. Nat. Prod. 2021, 84, 2104–2110. [Google Scholar] [CrossRef] [PubMed]
  114. Ying, Z.; Li, X.M.; Wang, B.G.; Li, H.L.; Meng, L.H. Rubensteroid A, a new steroid with antibacterial activity from Penicillium rubens AS-130. J. Antibiot. 2023, 76, 563–566. [Google Scholar] [CrossRef] [PubMed]
  115. He, Z.H.; Xie, C.L.; Hao, Y.J.; Xu, L.; Wang, C.F.; Hu, M.Y.; Li, S.J.; Zhong, T.H.; Yang, X.W. Solitumergosterol A, a unique 6/6/6/6/5 steroid from the deep-sea-derived Penicillium solitum MCCC 3A00215. Org. Biomol. Chem. 2021, 19, 9369–9372. [Google Scholar] [CrossRef] [PubMed]
  116. Youssef, D.T.A.; Shaala, L.A.; Almohammadi, A.; Elhady, S.S.; Alzughaibi, T.A.; Alshali, K.Z. Characterization of bioactive compounds from the Red Sea tunicate-derived fungus Penicillium commune DY004. Lett. Org. Chem. 2022, 19, 144–149. [Google Scholar] [CrossRef]
  117. Liu, X.Y.; Dong, Y.F.; Zhang, X.W.; Zhang, X.J.; Chen, C.X.; Song, F.H.; Xu, X.L. Two new trienoic acid derivatives from marine-derived fungus Penicillium oxalicum BTBU20213011. Rec. Nat. Prod. 2023, 17, 958–962. [Google Scholar]
  118. Yang, J.P.; Zhang, X.W.; Xu, W.; Xu, X.L.; Song, F.H. A new phenyl 6,7-dihydroxygeranyl ether derivative from a marine-derived fungus strain of Penicillium arabicum ZH3-9. Nat. Prod. Res. 2023. [Google Scholar] [CrossRef]
  119. Yang, S.Q.; Li, X.M.; Chen, X.D.; Li, X.; Wang, B.G. Three new α-pyrone derivatives from the soil-derived fungus Penicillium herquei MA-370. Nat. Prod. Res. 2023. [Google Scholar] [CrossRef]
  120. Newaz, A.W.; Yong, K.; Yi, W.W.; Wu, B.; Zhang, Z.Z. Antimicrobial metabolites from the indonesian mangrove sediment-derived fungus Penicillium chrysogenum sp. ZZ1151. Nat. Prod. Res. 2023, 37, 1702–1708. [Google Scholar] [CrossRef]
  121. Hsi, H.Y.; Wang, S.W.; Cheng, C.H.; Pang, K.L.; Leu, J.Y.; Chang, S.H.; Lee, Y.T.; Kuo, Y.H.; Huang, C.Y.; Lee, T.H. Chemical constituents and anti-angiogenic principles from a marine algicolous Penicillium sumatraense SC29. Molecules 2022, 27, 8940. [Google Scholar] [CrossRef] [PubMed]
  122. Liu, S.Z.; Tang, X.X.; He, F.M.; Jia, J.X.; Hu, H.; Xie, B.Y.; Li, M.Y.; Qiu, Y.K. Two new compounds from a mangrove sediment-derived fungus Penicillium polonicum H175. Nat. Prod. Res. 2022, 36, 2370–2378. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, W.H.; Zhang, J.W.; Qi, X.; Zhao, K.; Pang, X.Y.; Lin, X.P.; Liao, S.R.; Yang, B.; Zhou, X.F.; Liu, S.W.; et al. p-terphenyls as anti-HSV-1/2 agents from a deep-sea-derived Penicillium sp. J. Nat. Prod. 2021, 84, 2822–2831. [Google Scholar] [CrossRef] [PubMed]
  124. Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2023, 40, 275. [Google Scholar] [CrossRef] [PubMed]
  125. Hong, L.L.; Ding, Y.F.; Zhang, W.; Lin, H.W. Chemical and biological diversity of new natural products from marine sponges: A review (2009–2018). Mar. Life Sci. Technol. 2022, 4, 356–372. [Google Scholar] [CrossRef]
  126. Kozlovskii, A.G.; Zhelifonova, V.P.; Antipova, T.V. Fungi of the genus Penicillium as producers of physiologically active compounds (review). Appl. Biochem. Microbiol. 2013, 49, 1–10. [Google Scholar] [CrossRef]
Figure 1. Structures of 152.
Figure 1. Structures of 152.
Marinedrugs 22 00191 g001
Figure 2. Structures of 53105.
Figure 2. Structures of 53105.
Marinedrugs 22 00191 g002
Figure 3. Structures of 106153.
Figure 3. Structures of 106153.
Marinedrugs 22 00191 g003
Figure 4. Structures of 154191.
Figure 4. Structures of 154191.
Marinedrugs 22 00191 g004
Figure 5. Structures of 192246.
Figure 5. Structures of 192246.
Marinedrugs 22 00191 g005
Figure 6. Structures of 247296.
Figure 6. Structures of 247296.
Marinedrugs 22 00191 g006
Figure 7. Structures of 297364.
Figure 7. Structures of 297364.
Marinedrugs 22 00191 g007
Figure 8. Structures of 365402.
Figure 8. Structures of 365402.
Marinedrugs 22 00191 g008
Figure 9. Structures of 403408.
Figure 9. Structures of 403408.
Marinedrugs 22 00191 g009
Figure 10. Structures of 409445.
Figure 10. Structures of 409445.
Marinedrugs 22 00191 g010
Figure 11. The proportion of the Penicillium fungi derived from different marine habitats.
Figure 11. The proportion of the Penicillium fungi derived from different marine habitats.
Marinedrugs 22 00191 g011
Figure 12. The proportion of new bioactive compounds in each chemical class.
Figure 12. The proportion of new bioactive compounds in each chemical class.
Marinedrugs 22 00191 g012
Figure 13. The percentage distribution of new compounds with different bioactivities.
Figure 13. The percentage distribution of new compounds with different bioactivities.
Marinedrugs 22 00191 g013
Figure 14. (A) The proportion distribution of new compounds with different bioactivities in each chemical class; (B) the proportion distribution of new compounds with different chemical classes in each bioactivity.
Figure 14. (A) The proportion distribution of new compounds with different bioactivities in each chemical class; (B) the proportion distribution of new compounds with different chemical classes in each bioactivity.
Marinedrugs 22 00191 g014
Table 1. New compounds from marine-derived Penicillium fungi.
Table 1. New compounds from marine-derived Penicillium fungi.
No.CompoundsFungal Species/Strain No.Source of FungiBioactivitiesRef.
Azaphilones
1Penicilazaphilone IaP. sclerotiorum E23Y-1ASponge Cytotoxic activity
Anti-inflammatory
[17]
2Penicilazaphilone J
3epi-geumsanol D
45Penidioxolanes C–D
69Penicilazaphilone K–N
1011Penicilazaphilones F–GP. sclerotiorum E23Y-1ASponge Anti-inflammatory [22]
125-bromoisorotiorinP. sclerotiorum E23Y-1ASponge Antibacterial activity
Enzyme inhibitory
[18]
13Penicilazaphilone Ha
14Penicilazaphilone Hb P. sclerotiorumAlgae Anti-angiogenesis[20]
13Penicilazaphilone Ib
1611-epi-geumsanol F
1711-epi-geumsanol B
188a-epi-hypocrellone A P. sclerotiorumSediment Cytotoxic activity
Anti-inflammatory
[23]
198a-epi-eupenicilazaphilone C
20a/bIsochromophilone H (a/b) (isomers)P. sclerotiorum HY5Mangrove Phytotoxicity[24]
21Sclerotiorin A
22Sclerotiorin B
23Ochlephilone
24Isochromophilone IV
25a/bIsochromophilone J (a/b) (isomers)
2629Chermesinones D–GP. chermesinum FS625Seawater Anti-inflammatory[25]
3031Daldinins G–HP. glabrum glmu 003Soft coral Antibacterial activity
Enzyme inhibitory
[26]
Isocoumarins
3233Peniciisocoumarins I–JPenicillium sp. GXIMD 03001Mangrove Cytotoxic activity[27]
3435Penicillols A–BPenicillium sp. BJR-P2Mangrove Anti-inflammatory[28]
Chromones
3638Epiremisporines C–EP. citrinum BCRC 09F458Waste water Cytotoxic activity
Anti-inflammatory
[29]
3941Epiremisporines F–HP. citrinum BCRC 09F458Waste waterCytotoxic activity
Anti-inflammatory
[30]
4249Penithochromones M–TP. thomii Maire YPGA3Sediment Enzyme inhibitory
Antioxidant activity
[31]
5052Penithochromones U–WP. thomii YPGA3Sediment Enzyme inhibitory[32]
Citrinins
53(5R)-and (5S)-isoquinocitrinin B Penicillium sp. TW131-64Sediment Antibacterial activity [33]
54(5R)-and (5S)-isoquinocitrinin C
55(5R)-and (5S)-isoquinocitrinin D
56(3R,4S)-8-hydroxy-6-methoxy-3,4,5-trimethylisochromane-7-carboxylatemethyl
57(3R,4S)-6-hydroxy-8-methoxy-3,4,5-trimethylisochromane-7-carboxylatemethyl
58Penicitrinone J
5962Dicitrinone G–JPenicillium sp. GGF16-1-2Starfish Antifungal and cytotoxic activities[10]
6365Neotricitrinols A–CP. citrinum W23Sediment Anti-osteoporosis activity[34]
6667Xerucitrinins B–CP. citrinum Y34Sediment Enzyme inhibitory [35]
68Penicitrinol PPenicillium sp. SCSIO 41302Sponge Antibacterial activity
Enzyme inhibitory
[36]
69Dicitrinol DPenicillium sp. SCSIO 41303Sponge Antibacterial, cytotoxic, antiviral activities, and enzyme inhibitory[37]
β-resorcylic acids
7014-hydroxyasperentin BP. antarcticum KMM 4685Brown alga Cytotoxic activity[38]
7173β-resoantarctines A–C
748-dehydro-β-resoantarctine A
Verrucosidins
75769-O-ethylpenicyrones A–B P. cyclopium SD-413Sediment Antibacterial activity[40]
7782Poloncosidins A–FP. polonicum CS-252Sediment Antibacterial activity[39]
8387Poloncosidins G–KP. polonicum CS-252Sediment Antibacterial activity[41]
8889Verrucosidinol A–BP. griseofulvum MCCC 3A00225Sediment Anti-food allergy[42]
Citreoviridins
9091Citreoviridins H–IPenicillium sp. BJR-P2Mangrove Anti-inflammatory[27]
9297Citreoviridins J–OP. citreonigrum MCCC 3A00169Sediment Cytotoxic activity
Anti-inflammatory
[43]
Nitrogen-containing polyketides
9899Oxopyrrolidines A–BP. oxalicumMEFC104Sediment Antibacterial activity[44]
1007-hydroxy-3,10-dehydrocyclopeptineP. polonicum MCCC3A00951Sediment Antiviral activity[45]
101105Steckfusarins A–EP. steckii SCSIO41040Green algaeAntibacterial, antifungal, cytotoxic and antiviral activities
Enzyme inhibitory Antioxidant
Anti-inflammatory
[46]
Sorbicillinoids
106108Bisorbicillchaetones A–CPenicillium sp. SCSIO06868Sediment Anti-inflammatory[47]
10910-Methylsorbiterrin APenicillium sp. SCSIO06871Sediment Antibacterial and antifungal activities
Enzyme inhibitory
[48]
110Epitetrahydrotrichodimer ether
111Demethyldihydrotrichodimerol
112Bisorbicillpyrone A
113Dihydrotrichodermolidic acid
1145-hydroxy-dihydrodemethy lsorbicillin
115Sorbicillpyrone A
1165,6-dihydrovertinolide
117118Sorbicatechols C–DP. allii-sativi MCCC3A00580Seawater Cytotoxic activity[49]
119(4E)-1-(4,6-Dihydroxy-5-methylpyridin-3-yl)hex-4-en-1-onePenicillium sp. DM815Mangrove Anti-inflammatory[50]
Isochromans
120125Penicisteckins A–FP. steckii HNNU-5B18Beach mudAntibacterial activity
Cytotoxic activity
[51]
α-pyrone polyketides
126130Penipyrols C-GPenicillium sp. HDN-11-131Mangrove Cytotoxic activity[52]
131Methyl-penipyrol A
Hirsutellones
132Perpyrrospirone AP. citrinumSeawater Cytotoxic activity[53]
Xanthones and benzophenones
133
134
11-O-acetylaspergillusone BPenicillium sp. MCCC 3A00126Sediment Cytotoxic activity
Ferroptosis inhibitory
[54]
7-dehydroxyhuperxanthone A
135Penicixanthene EPenicillium sp. GXIMD 03101Mangrove Cytotoxic activity[55]
136Penibenzophenone CPenicillium sp.Mangrove Antibacterial and insecticidal activities[16]
Hydroxybenzenes
137Peniketide APenicillium sp. SCZ-1Sediment Enzyme inhibition[56]
138Methyl ester of penipyrol A
139141Penidihydrocitrinins A–CP. citrinum W17Sediment Anti-inflammatory
Anti-osteoporosis
[57]
142Peniciphenalenin GP. oxalicumSeawaterCytotoxic activity[58]
143Penicinone CPenicillium sp. LA032Mangrove ----[59]
1445-((R,1Z,3E)-6-hydroxy-1,3-heptadien-1-yl)-1,3-benzenediolPenicillium sp. TW58-16Sediment Anti-inflammatory
Enzyme inhibition
[60]
1454-carboxy-5-((R,1Z,3E)-6-hydroxy-1,3-heptadien-1-yl)-1,3-benzenediol
1464-carboxy-5-((1Z,3E)-1,3-heptadien-1-yl)-1,3-benzenediol
1475-((1Z,3E)-4-carboxy-1,3-butadienyl-1-yl)-1,3-benzenediol
148(2E)-3-[(3R)-3,4-dihydro-6,8-dihydroxy-1-oxo-1H-2-benzopyran-3-yl]-2-propenoic acid
1493-[(3S)-3,4-dihydro-6,8-dihydroxy-1-oxo-1H-2-benzopyran-3-yl]-propanoic acid
150Coniochaetone NPenicillium sp. SCSIO06868Sediment Antibacterial activity[61]
Lactones
151152Penicinones A–BPenicillium sp. LA032Mangrove Cytotoxic activity[59]
153Walterolactone EPenicillium sp. TW58-16Sediment Antibacterial activity[62]
Olefinic acids and their derivatives
154157Steckwaic acid A-DP. steckii AS-324Coral Antibacterial and antifungal activities[64]
15811-ketotanzawaic acid D
1596,15-dihydroxytanzawaic acid M
16015R-methoxytanzawaic acid M
16115S-methoxytanzawaic acid M
1628-hydroxytanzawaic acid M
1638-hydroxytanzawaic acid B
164168Steckwaic acid Ea–IaP. steckii AS-324Coral Antibacterial activity[19]
16918-O-acetyltanzawaic acid R
17010-hydroxytanzawaic acid U
17113R-tanzawaic acid S
172176Steckwaic acid Eb–IbP. steckii SCSIO 41040Green algae Antibacterial, antifungal, cytotoxic, and antiviral activities[63]
177178Steckwaic acid J–K
179182Penicisteck acid A–DP. steckii SCSIO 41025MangroveAntibacterial activity
Enzyme inhibition
[65]
183186Penifellutins A–DP. crustosum PRB-2 and P. fellutanum HDN14-323SeawaterCytotoxic activity[66]
Other polyketides
187Rubenpolyketone AP. rubens AS-130Coral Antibacterial activity[67]
188Oxalichroman AP. oxalicum 2021CDF-3Red algae Cytotoxic activity[68]
189Oxalihexane A
190Leptosphaerone DPenicillium sp. TW58-16Sediment Antibacterial activity[62]
19115-O-methyl ML-236AP. solitum MCCC 3A00215Sediment Cytotoxic activity
Anti-food allergy
[69]
Indole alkaloids
192195Communesins M–PP. expansum MMS42Sediment Cytotoxic and neuroprotective activities[9]
196Deoxy-14,15-dehydroisoaustamideP. dimorphosporum KMM 4689Soft coralCytotoxic activity[70]
19716α-hydroxy-17β-methoxy-deoxydihydroisoaustamideP. dimorphosporum KMM 4689Soft coral Cytotoxic and neuroprotective activities[71]
19816β-hydroxy-17α-methoxy-deoxydihydroisoaustamide
19916β,17α-dihydroxy-deoxydihydroisoaustamide
20016α-hydroxy-17α-methoxy-deoxydihydroisoaustamide
20116α,17α-dihydroxy-deoxydihydroisoaustamide
20216,17-dihydroxydeoxydihydroisoaustamide
2033β-hydroxy-deoxyisoaustamide
204Penilline DPenicillium sp. SCSIO 05705Soil Antibacterial and cytotoxic activities
Enzyme inhibition
[72]
205Penindolacid APenicillium sp. LW92Sediment Antioxidant activity
Enzyme inhibitory
[73]
206207Penicamides A–BPenicillium sp. LA032Soil Cytotoxic activity[59]
20811S-(−)-penilloid APenicillium sp. ZZ1750Marine mud Cytotoxic activity[74]
20911R,14E-(+)-penilloid A
Pyridones
210220Penicipyridones A–KP. oxalicum QDU1Leaves of plantAnti-inflammatory[75]
Quinolinones
221PenicinolonePenicillium sp. SCSIO 41033Sponge Antibacterial and antifungal activities
Enzyme inhibitory
[76]
222Quinolactone AP. citrinum YX-002Mangrove Enzyme inhibitory[77]
223Quinolactacin C1
2243-epi-quinolactacin C1
225a/bQuinolactacin E (a racemic mixture)Penicillium sp. SCSIO 41303Sponge Cytotoxic and antiviral activities
Enzyme inhibitory
[37]
226Quinolactacin F1
227Quinolactacin F2
228a/bQuinolactacin G (enantiomers)
229232(±)-oxypenicinolines A–D P. steckii SCSIO 41025Mangrove Antibacterial, antifungal, and cytotoxic activities
Enzyme inhibitory
[78]
(racemic mixtures, respectively)
233234Penicinoline F–G
Decahydrofluorene-class alkaloids
235241Pyrrospirone K–QPenicillium sp. SCSIO 41512Soft coral Antibacterial and cytotoxic activities
Enzyme inhibitory
[79]
Piperazines
242Adametizine CP. ludwigii SCSIO 41408SedimentAntibacterial, antifungal, and cytotoxic activities
Anti-osteoporosis
[80]
243(8S,9R,12R,18S)-12-hydroxy-fumitremorgin BPenicillium sp. TW58-16Sediment Antibacterial activity[62]
244246Penigainamides A–CP. steckii YESeawater Cytotoxic activity[81]
Tetramic-acid-based alkaloids
247249Tolypocladenols D–FP. oxalicum QDU1Leaves of plantAntifungal and cytotoxic activities
Anti-inflammatory
[75]
250251Penicillenols G1–G2Penicillium sp. SCSIO06868Sediment Antibacterial and antiviral activities[61]
252Penicillenol H
Amines and amides
253(Z)-4-(5-acetoxy-N-hydroxy-3-methylpent-2-enamido) butanoateP. oxalicum HLLG-13Mangrove Antibacterial and insecticidal activities[15]
254255Polonimides D–E Penicillium sp. SCSIO 41413Sponge Antibacterial and cytotoxic activities
Anti-inflammatory
[82]
256Speradine IPenicillium sp. SCSIO 41038Soft coral Cytotoxic activity
Enzyme inhibitory
[83]
257(S)-2-acetamido-4-(2-(methylamino)phenyl)-4-oxobutanoic acidP. citrinum XIA-16Shrimp Ferroptosis inhibitory [84]
258Citrinadin CP. citrinumSediment Antibacterial and cytotoxic activities[85]
259(2S,2′R,3R,3′E,4E,8E)-N-2′-hydroxyhexadecanoyl-2-amino-9-methyl-4,8-octadecadiene-1,3-diolP. chrysogenum Y20-2Seawater Anti-angiogenesis [86]
260261Penichryfurans A–BP. chrysogenumRed alga Cytotoxic activity[87]
262280Talaroenamines F1−F19P. malacosphaerulum HPU-J01Wetland Cytotoxic activity[88].
281PeniokaraminePenicillium sp. LSH-3-1Sediment Cytotoxic activity
Anti-inflammatory
[89]
282Penipyranopyridine
283284Penicidihydropyridones A–BPenicillium sp. B9Sponge Cytotoxic activity[90]
285(+)-solitumidine DP. solitum MCCC 3A00215Sediment Cytotoxic activity
Anti-food allergy
[69]
286(±)-solitumidine E (a racemic mixture)
287Penicmariae-crucis C acidP. steckii SCSIO 41025Mangrove Antibacterial and antifungal activities
Enzyme inhibitory
[65]
288N-(6-hydroxy-2-oxoindolin-3-ylidene)-5′-methoxy-5′-oxobutyl-amine oxide
289Methyl-1′-(N-hydroxyacetamido)-butanoate
290PenigrisamideP. griseofulvum MCCC 3A00225Sediment Anti-food allergy[42]
291Aurantiomoate C
292N,N-pyroglutamylleucinmethylester
293Methyl 2S-hydroxy-3-methylbutanoyl-L-leucinate
2946R,7-dihydroxy-3,7-dimethyloctanamide
Other alkaloids
295SulfoxanthocillinPenicillium sp. SCSIO sof101Seawater Antibacterial activity
Anti-inflammatory
[91]
296Penipyridinone BPenicillium sp. ZZ1750Sea mud Cytotoxic activity[74]
Sesquiterpenes
297Chermesiterpenoid DP. rubens AS-130Coral Antibacterial activity[67]
298307Copteremophilanes A–JP. CopticolaSponge Cytotoxic activity
Neuroprotection
[92]
308Astellolide Q Penicillium sp. N-5Soil Antibacterial and antifungal activities[14]
309Chrysoride AP. chrysogenum LD-201810Red algaCytotoxic activity[93]
310Purpuride DPenicillium sp. ZZ1283Sea mudAntibacterial activity[94]
311328Bilaiaeacorenols A–RP. bilaiae F-28Sediment Anti-inflammatory[95]
329331Citreobenzofurans D–FPenicillium sp. HDN13-494Soil Antibacterial and cytotoxic activities[96]
332333Phomenones A–B
334(2S,3S,5S,6S,7S,8R,11S,12R)-15-deacetyl-7,8-dihydroxycalonectrinPenicillium sp. LXY140-R and Penicillium sp. LXY140-3Sediment Cytotoxic activity [97]
3351-Methyl-4-[3,4,5-trihydroxy-1,2,2-trimethylcyclopently]benzene
336a/b(±)Methylsulfinyl-1-hydroxyboivinianin A (enantiomers)P. chrysogenum LD-201810Red algaAntifungal and cytotoxic activities[98]
337(4S,5R,9S,10R)-11,13-dihydroxy-drim-7-en-6-onePenicillium sp. TW58-16Sediment Anti-inflammatory
Enzyme inhibition
[60]
338(4S,5R,9S,10R)-11-hydroxy-13-carboxy-drim-7-en-6-one
Diterpenes
339Penijanthine EP. citrinum ZSS-9Sediment Antiviral activity[99]
340348Janthinellumines A–IP. janthinellumSeawater Antibacterial and antiviral activity
Enzyme inhibition
[100]
349350Oxalierpenes A–BP. oxalicumShrimp Antiviral activity[101]
3514-hydroxyleptosphin CP. antarcticum KMM 4670SedimentAntibacterial activity
Enzyme inhibition
[102]
35213-epi-Conidiogenone F
353355Shearinines R–TPenicillium sp. UJNMF0740SedimentAntibacterial activity
Neuroprotection
[103]
35622-hydroxyshearinine I
357358Conidiogenones J–KP. oxalicum HLLG-13Mangrove Antibacterial and insecticidal activities[15]
359362Penerpenes K–NPenicillium sp. KFD28Mollusk Antibacterial and cytotoxic activities[104]
363EpipaxillinePenicillium sp. KFD28MolluskEnzyme inhibition[105]
364Penerpene J
Meroterpenes
365371Peniscmeroterpenoids H–NP. sclerotiorum GZU-XW03-2Mollusk Anti-inflammatory[106]
372Andrastin IP. ochrochloronSeawater Antibacterial activity[107]
373376Chermesin E–HP. chermesinum EN-480Red alga Antibacterial activity[108]
377383Peniscmeroterpenoid A–GP. sclerotiorum GZU-XW03-2Mollusk Anti-inflammatory[109]
384Meroterpenthiazole AP. allii-sativi MCCC 3A00580SeawaterCytotoxic activity[110]
385Citrehybridonol BP. allii-sativi MCCC 3A00580SeawaterAnti-allergic bioactivity[111]
386Andrastin G
387393Andrastones B–H
394396Hemiacetalmeroterpenoids A–CPenicillium sp. N-5Soil Antifungal activity[14]
397399Penimeroterpenoids A–CPenicillium sp.Sediment Cytotoxic activity[112]
400402Penicianstinoids C–EPenicillium sp. TGM112Mangrove Antifungal and insecticidal activities[113]
Steroids
403Rubensteroid AP. rubens AS-130Coral Antibacterial activity[114]
404Andrastin HP. oxalicum HLLG-13Mangrove Insecticidal activity[15]
405Solitumergosterol AP. solitum MCCC 3A00215Sediment Cytotoxic activity[115]
Peptides
406407Penicamides A–BPenicillium sp. SCSIO 41512Soft coral Antifungal activity[21]
408Penicillizine AP. commune DY004Tunicate Cytotoxic activity[116]
Others
409410Penioxa acids A–BP. oxalicum BTBU20213011Sediment Antibacterial and antifungal activities[117]
411(Z)-5-acetoxy-3-methylpent-2-enoic acidP. oxalicum HLLG-13Mangrove Antibacterial and insecticidal activities[15]
412Antaketide AP. antarcticum KMM 4670SedimentAntibacterial activity[102]
413Ochrochloronic acidP. ochrochloronSea sandAntibacterial and cytotoxic activities[107]
414(Z)-4-((6,7-dihydroxy-3,7-dimethyloct-2-en-1-yl)oxy)benzoic acidP. arabicum ZH3-9Sea mud Antibacterial and antifungal activities[118]
415417Annularin L–NP. herquei MA-370Soil Antibacterial activity[119]
418Peniprenylphenol AP. chrysogenum ZZ1151Sediment Antibacterial activity[120]
41913-(11-hydroxy-8-(4-hydroxy-1,6-dimethoxybenzyl)-9-methoxy-12-methylphenyl) propan-15-oneP. steckii SCSIO 41040Green algaeAntibacterial, antifungal, cytotoxic and antiviral activities Anti-inflammatory[63]
420Speradine JPenicillium sp. SCSIO 41038Soft coral Cytotoxic activity
Enzyme inhibitory
[83]
421Eutypoid FPenicillium sp. SCSIO 41413Sponge Antibacterial activity[82]
422Penisterines AP. sumatraense SC29Alga Anti-angiogenesis [121]
423425Penisterines C–E
426Penisterine A methyl ether
4272-methyl-3-(5-oxohexyl) maleic acidP. ludwigii SCSIO 41408Sediment Antibacterial, antifungal, and cytotoxic activities
Anti-osteoporosis
[80]
4282-(4-hydroxyhexyl)-3-methylmaleic acid
4293-(ethoxycarbonyl)-2-methylenenonanoic acid
4307-hydroxy-3-(methoxycarbonyl)-2-methylenenonanoic acid
4312-(4-hydroxypentyl)-4-methyl-5-oxo-2,5-dihydrofuran-3-carboxylic acid
4325,6-Dihydroxy-3-methoxyhex-2-enoic acidPenicillium sp. LXY140-R and Penicillium sp. LXY140-3Sediment Cytotoxic activity[97]
4336-acetyl-4-methoxy-3,5-dimethyl-2H-pyran-2-oneP. polonicum H175Sediment Hypoglycemic effect[122]
434(2E,4E)-5-((2S,3S,4R,5R)-3,4-dihydroxy-2,4,5-trimethyltetrahydrofuran-2-yl)-2,4-dimethylpenta-2,4-dienal
4355-glycopenostatin FP. CopticolaSponge ___[92]
4365-glucopenostatin I
437a/b(±)-TetraketidePenicillium sp. SCSIO 41302Sponge Antibacterial and cytotoxic activities
Enzyme inhibitory
[36]
4388-hydroxyhelvafuranoneP. griseofulvum MCCC 3A00225Sediment Anti-food allergy [42]
439Methyl-3,7,9-trihydroxydecanate
4409-hydroxy-3,7-epoxydecanoic acid
441442Chrysoalides A–B P. chrysogenum LD-201810Red alga Antifungal and cytotoxic activities[98]
443445Peniterphenyls A–CPenicillium sp. SCSIO41030Sediment Antiviral activity
Enzyme inhibitory
[123]
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

Lv, F.; Zeng, Y. Novel Bioactive Natural Products from Marine-Derived Penicillium Fungi: A Review (2021–2023). Mar. Drugs 2024, 22, 191. https://doi.org/10.3390/md22050191

AMA Style

Lv F, Zeng Y. Novel Bioactive Natural Products from Marine-Derived Penicillium Fungi: A Review (2021–2023). Marine Drugs. 2024; 22(5):191. https://doi.org/10.3390/md22050191

Chicago/Turabian Style

Lv, Fang, and Yanbo Zeng. 2024. "Novel Bioactive Natural Products from Marine-Derived Penicillium Fungi: A Review (2021–2023)" Marine Drugs 22, no. 5: 191. https://doi.org/10.3390/md22050191

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