Marine DrugsPub Date : 2025-03-23DOI: 10.3390/md23040139
Felix Kerlikowsky, Madeline Bartsch, Wiebke Jonas, Andreas Hahn, Jan Philipp Schuchardt
{"title":"Calanus Oil and Lifestyle Interventions Improve Glucose Homeostasis in Obese Subjects with Insulin Resistance.","authors":"Felix Kerlikowsky, Madeline Bartsch, Wiebke Jonas, Andreas Hahn, Jan Philipp Schuchardt","doi":"10.3390/md23040139","DOIUrl":"https://doi.org/10.3390/md23040139","url":null,"abstract":"<p><p>Obesity increases the risk for insulin resistance (IR) and type-2 diabetes. Lifestyle interventions (LI) and dietary supplementation can help mitigate IR. We investigated the effect of calanus oil (CO) supplementation, combined with LI, on glucose homeostasis in obese subjects. CO, a novel marine oil, contains omega-3 fatty acid wax esters as well as plant sterols and astaxanthin. In the double-blind, randomized, placebo-controlled 12-week intervention study, 266 subjects with distinct IR phenotypes were assigned to four groups: 2 g CO/day, 4 g CO/day, 2 g CO/day + LI, and placebo. The effect of CO on HOMA index reduction was influenced by the initial (t<sub>0</sub>) squared HOMA index (interaction <i>p</i> = 0.011). A post hoc test showed significant improvement with 2 g CO/day + LI (estimated marginal means [EMM] 95% confidence interval [CI]: -0.19 [-0.80-0.41], <i>p</i> = 0.021). Secondary analysis revealed that 4 g CO/day had significant effects in subjects with mild IR (HOMA index 2.5-5.0) (EMM [95% CI]: -0.76 [-1.53-0.03], <i>p</i> = 0.043). CO supplementation improved glucose homeostasis, with effects varying by dose, combination with LI, and IR phenotype.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 4","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12028837/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144034011","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marine DrugsPub Date : 2025-03-22DOI: 10.3390/md23040138
Jin-Hyung Kim, Jung-Rae Rho, Jung-Hye Choi
{"title":"Marine Sponge-Derived Gukulenin A Sensitizes Ovarian Cancer Cells to PARP Inhibition via Ferroptosis Induction.","authors":"Jin-Hyung Kim, Jung-Rae Rho, Jung-Hye Choi","doi":"10.3390/md23040138","DOIUrl":"https://doi.org/10.3390/md23040138","url":null,"abstract":"<p><p>Resistance to PARP inhibitors (PARPi), such as olaparib (OLA), is a major challenge in ovarian cancer treatment. In this study, we investigated the combination effect of PARPi and gukulenin A (GUA), a bis-tropolone tetraterpenoid isolated from the marine sponge <i>Phorbas gukhulensis</i>. We found that GUA at a mildly cytotoxic dose synergistically enhanced OLA-induced cytotoxicity in human ovarian cancer cells. The combination treatment significantly increased reactive oxygen species (ROS) levels and lipid peroxidation, leading to ferroptotic rather than apoptotic cell death. Network pharmacology and gene ontology (GO) enrichment analyses revealed oxidative stress-related pathways as key mediators of this effect. Inhibition of NADPH oxidase (NOX) reversed combination-induced cell death, while ferrostatin-1 (FER-1), a ferroptosis inhibitor, significantly reduced lipid peroxidation and cytotoxicity. Additionally, GUA and OLA treatment suppressed ERK1/2 activation, and ERK overexpression attenuated the combination-induced cell death. Collectively, these findings suggest that marine-derived GUA enhances PARPi efficacy in ovarian cancer cells by inducing ferroptosis through oxidative stress and ERK pathway modulation.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 4","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12028354/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143971301","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Marine-Derived Yaequinolone Derivative CHNQD-02792 Suppresses Colorectal Cancer Cell Proliferation and Induces Apoptosis via MAPK Pathway Modulation.","authors":"Jia-Qi Kang, Tian-Yi Zhou, Wen-Hui Wang, Mei-Yan Wei, Chang-Lun Shao","doi":"10.3390/md23040136","DOIUrl":"https://doi.org/10.3390/md23040136","url":null,"abstract":"<p><p>Colorectal cancer is currently the third most common malignancy, and the toxic side effects of clinical therapeutic drugs often influence treatment outcomes. Marine-derived quinolone alkaloids exhibit various biological activities and are particularly notable for their antitumor properties. Compounds <b>1</b>-<b>13</b> were semi-synthesized based on 4'-desmethoxyyaequinolone J1, which is a 4-phenyl derivative of the natural quinolone alkaloid yaequinolone J1 and was isolated from <i>Penicillium</i> sp. FKI-2140. This study is the first to investigate the antitumor activity of <b>1</b>-<b>13</b> in colorectal cancer cells through proliferation, clonality, apoptosis, cell cycle, and MAPK signaling pathway. Cytotoxicity screening against seven colorectal cancer cell lines revealed that CHNQD-02792 (<b>13</b>) had the most sensitivity to HT-29 cells (IC<sub>50</sub> = 4.5 μM), far exceeding positive control 5-fluorouracil (IC<sub>50</sub> = 15.58 μM). The plate cloning assay revealed that CHNQD-02792 completely inhibited the growth of HT-29 cells at the concentration of 9 μM. CHNQD-02792 (4.5 μM) inhibited CDK1 expression and triggered G2/M phase arrest in HT-29 cells. Mechanistic analysis revealed that CHNQD-02792 induced apoptosis by suppressing the anti-apoptotic protein Bcl-2 and upregulating the pro-apoptotic proteins Caspase-3 and Bax. Furthermore, CHNQD-02792 inhibited ERK and JNK phosphorylation and thus highlighted its regulatory role in MAPK signaling. These findings suggest that CHNQD-02792 exerts cytotoxic effects on HT-29 cells via dual mechanisms: inducing G2/M arrest and apoptosis while regulating MAPK signaling through ERK/JNK dephosphorylation. This study demonstrates the dual targeting of CHNQD-02792 against tumor cell proliferation and survival pathways, providing a foundation for further development of anti-colorectal cancer drugs.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 4","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12028472/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144023392","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Discovery of PD-L1 Peptide Inhibitors from Ascidian Enzymatic Hydrolysates by Affinity Ultrafiltration Coupled to NanoLC-MS/MS.","authors":"Qiuyang Huang, Xiaoling Zang, Xinyu Jin, Qian Liu, Xin Zhang, Xinyu Li, Lizhen Zhao, Zhihua Lv","doi":"10.3390/md23040137","DOIUrl":"https://doi.org/10.3390/md23040137","url":null,"abstract":"<p><p>Anti-PD-1 and anti-PD-L1 antibodies have achieved great clinical success in cancer immunotherapy, and peptide and small molecule inhibitors of PD-1/PD-L1 binding also attract much attention. Ascidians are not only seafood, but are also an important source of bioactive substances, including anti-tumor components. In this study, ascidian enzymatic hydrolysates were found to contain PD-1/PD-L1 inhibitory components. Affinity ultrafiltration (AUF) coupled with the nanoLC-MS/MS method was first applied in screening for PD-L1 peptide inhibitors from ascidian enzymatic hydrolysates. Two anti-PD-L1 ascidian peptides, C5 (LDVVIHTVTYGDR) and S2 (VLRDNIQGITKPAIR), were filtered out from the ascidians <i>Ciona intestinalis</i> and <i>Styela clava</i>, respectively. C5 and S2 showed moderate anti-PD-1/PD-L1 effects with the IC<sub>50</sub> values of 33.9 µM (C5) and 112.8 μM (S2), respectively, by homogenous time-resolved fluorescence (HTRF) binding assay, and the K<sub>D</sub> values of 22.9 µM (C5) and 29.1 µM (S2), respectively, by surface plasmon resonance (SPR) assay. The results of this study suggest that ascidian enzymatic hydrolysates may be a potential source of bioactive peptides with anti-PD-1/PD-L1 activity.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 4","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12028738/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144006953","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marine DrugsPub Date : 2025-03-20DOI: 10.3390/md23030135
Jeong Won Ahn, Hyun Soo Kim, So Hui Kim, Hye Soo Yang, Kongara Damodar, Yeong-Min Yoo, Jin Tae Hong, Seong Soo Joo
{"title":"Amelioration of Particulate Matter-Induced Oxidative Stress by a Bioactive <i>Hizikia fusiformis</i> Extract: A Functional Biomaterial for Cosmeceutical Applications.","authors":"Jeong Won Ahn, Hyun Soo Kim, So Hui Kim, Hye Soo Yang, Kongara Damodar, Yeong-Min Yoo, Jin Tae Hong, Seong Soo Joo","doi":"10.3390/md23030135","DOIUrl":"10.3390/md23030135","url":null,"abstract":"<p><p>Air pollution-related skin damage has heightened the demand for natural protective agents. <i>Hizikia fusiformis</i>, a brown seaweed rich in fucoidan and bioactive fatty acids (α-linolenic acid, eicosatetraenoic acid, and palmitic acid), possesses antioxidant and anti-inflammatory properties. This study investigated the protective effects of <i>H. fusiformis</i> ethanol extract (HFE) against particulate matter (PM)-induced oxidative stress, inflammation, and apoptosis in human keratinocytes. Antioxidant activity was assessed using DPPH and hydroxyl radical scavenging assays, while PM-induced cytotoxicity, ROS generation, inflammatory markers, and apoptotic pathways were evaluated using the WST-8 assay, DCFH2-DA, qPCR, western blotting, and Hoechst staining. HFE significantly reduced ROS levels, enhanced antioxidant enzyme activity, and mitigated PM-induced cytotoxicity. These effects were mediated by fucoidan and fatty acids, which modulated inflammatory pathways (NF-κB and MAPK), stabilized membranes, and inhibited apoptosis (Bcl-2, Bax, and caspase-3). Collectively, these findings highlight HFE's potential as a natural anti-pollution skincare ingredient, supporting further in vivo studies and formulation development.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 3","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11943920/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143710566","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"<i>Thraustochytrium</i> sp. and <i>Aurantiochytrium</i> sp.: Sustainable Alternatives for Squalene Production.","authors":"Júnior Mendes Furlan, Graciela Salete Centenaro, Mariane Bittencourt Fagundes, Carlos Borges Filho, Irineu Batista, Narcisa Bandarra","doi":"10.3390/md23030132","DOIUrl":"10.3390/md23030132","url":null,"abstract":"<p><p>This study investigated a sustainable alternative to squalene production utilizing <i>Thraustochytrium</i> sp. and <i>Aurantiochytrium</i> sp., thereby reducing dependence on critically endangered sharks exploited for this compound. By optimizing fed-batch cultivation, a technique prevalent in industrial biotechnology, we have enhanced squalene yields and have demonstrated, through sensitivity analysis, the significance of this shift in preserving species at risk of extinction. Optimization of culture conditions led to the highest biomass concentrations for <i>Thraustochytrium</i> sp. being achieved at lower C-N ratios (<5.0), while the optimal biomass production for <i>Aurantiochytrium</i> sp. occurred in culture media with a high C-N ratio of 54:50. Regarding squalene production, <i>Thraustochytrium</i> sp. produced 26.13 mg/L in the fed-batch system after 72 h, and <i>Aurantiochytrium</i> sp. produced 54.97 mg/L in a batch system with 30 g/L glucose and 0.22 g/L nitrogen after 96 h, showcasing their potential for industrial applications. Moreover, the sensitivity analysis revealed that, on an industrial scale, both strains could produce up to 59.50 t of squalene annually in large-scale facilities, presenting a valuable and sustainable alternative for the biotechnological industry and significantly reducing the reliance on non-renewable and endangered sources such as shark liver oil and preventing the annual capture of over 156,661 sharks.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 3","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11944157/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143710563","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Marine-Derived Alternariol Suppresses Inflammation by Regulating T Cell Activation and Migration.","authors":"Chenfeng Liu, Fudie Gu, Zhengbiao Zou, Fengli Wang, Dashuai Li, Jing Song, Yazhen Hong, Xuhui Wu, Xianwen Yang, Wen-Hsien Liu, Guangming Liu, Yu Zhou, Qingmei Liu","doi":"10.3390/md23030133","DOIUrl":"10.3390/md23030133","url":null,"abstract":"<p><p>T cells play pivotal roles in inflammation's initiation and progression. Exploring natural compounds that regulate T cell function is crucial for preventing and treating inflammation. Herein, we report that Alternariol (AOH), a marine-derived secondary metabolite, exerts an anti-inflammatory activity by targeting T cell function. Using an ovalbumin (OVA)-induced OT-II CD4<sup>+</sup> T cell activation model, we demonstrated that AOH potently suppresses T cell proliferation and cytokine secretion, mildly promotes T cell apoptosis, and spares antigen presentation processes. Mechanistically, AOH controlled early T cell activation by inhibiting the expression of activation markers (CD69, CD25, CD44) and transcription factors (T-bet, Eomes), leading to impaired Th1 cytokine production. In vivo experiments revealed that AOH attenuated OVA-induced lung injury in mice by reducing immune cell infiltration in pulmonary tissues and draining lymph nodes. Notably, AOH dramatically suppressed OVA-specific T cells migrating to the inflammatory lung while impairing T-cell-mediated other immune cell infiltration. Collectively, AOH exhibited potent anti-inflammatory effects by modulating T cell proliferation, function, and migration, offering a promising therapeutic strategy for T-cell-mediated inflammatory diseases.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 3","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11944012/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143710509","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marine DrugsPub Date : 2025-03-19DOI: 10.3390/md23030134
Yanglong Liu, Jiawei Sun, Qihao Yan, Bingjian Wen, Yan Bai, Qishi Che, Hua Cao, Jiao Guo, Zhengquan Su
{"title":"Chitosan Oligosaccharides Prevent Alcohol-Induced Liver Disease by Attenuating Inflammation and Oxidative Stress.","authors":"Yanglong Liu, Jiawei Sun, Qihao Yan, Bingjian Wen, Yan Bai, Qishi Che, Hua Cao, Jiao Guo, Zhengquan Su","doi":"10.3390/md23030134","DOIUrl":"10.3390/md23030134","url":null,"abstract":"<p><p>Alcoholic liver disease (ALD) is a liver disorder resulting from excessive alcohol intake, and currently, there are no therapeutics approved by the FDA for its treatment. This study investigates the protective effects and underlying pharmacological mechanisms of two chitosan oligosaccharides, COST (MW ≤ 1000 Da) and COSM (MW ≤ 3000 Da), in mitigating alcohol-induced liver disease (ALD). In animal models, we evaluated the changes in ALD following treatment with COST and COSM. Histopathological analysis revealed that both COST and COSM interventions mitigated hepatic steatosis and inflammatory infiltration. Additionally, these compounds reduced various markers of liver injury, enhanced antioxidant enzyme levels, and significantly improved liver function. Western blot analysis demonstrated that COSM markedly decreased the expression of the hepatic metabolic enzyme CYP2E1, activated the Keap-1/Nrf-2/HO-1 pathway, and restrained the NF-κB and MAPK pathways. In an in vitro model of alcohol-induced hepatocyte L02 injury, both COST and COSM exhibited protective effects on hepatocytes, corroborating the findings from the animal studies. Collectively, in vivo and in vitro experiments confirmed that COST and COSM can reduce oxidative damage, enhance antioxidant capacity, and ameliorate steatosis and inflammatory damage in the liver, thereby significantly attenuating alcohol-induced injury. Notably, COSM exhibited slightly superior efficacy compared to COST.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 3","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11943892/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143710307","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Genome Mining-Guided Discovery of Two New Depsides from <i>Talaromyces</i> sp. HDN1820200.","authors":"Xiao Zhang, Luyang Liu, Jiani Huang, Xingtao Ren, Guojian Zhang, Qian Che, Dehai Li, Tianjiao Zhu","doi":"10.3390/md23030130","DOIUrl":"10.3390/md23030130","url":null,"abstract":"<p><p>Depsides and their derivatives are a class of polyketides predominantly found in fungal extracts. Herein, a silent nonreducing polyketide synthase (TalsA)-containing gene cluster, which was identified from the Antarctic sponge-derived fungus <i>Talaromyces</i> sp. HDN1820200, was successfully activated through heterologous expression in <i>Aspergillus nidulans</i>. This activation led to the production of two novel depsides, talaronic acid A (<b>1</b>) and B (<b>2</b>), alongside three known compounds (<b>3</b>-<b>5</b>). The further co-expression of TalsA with the decarboxylase (TalsF) demonstrated that it could convert <b>2</b> into its decarboxylated derivative <b>1</b>. The structural elucidation of these compounds was achieved using comprehensive 1D and 2D-NMR spectroscopy, which was complemented by HR-MS analysis. Talaronic acids A and B were firstly reported heterodimers of 3-methylorsellinic acid (3-MOA) and 5-methylorsellinic acid (5-MOA). All isolated compounds (<b>1</b>-<b>5</b>) were tested for their anti-inflammatory potential. Notably, compounds <b>1</b> and <b>2</b> exhibited anti-inflammatory activity comparable to that of the positive control. These results further enrich the structural class of depside natural products.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 3","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11944171/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143710424","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Diterpenoids of Marine Organisms: Isolation, Structures, and Bioactivities.","authors":"Qi Shi, Shujie Yu, Manjia Zhou, Peilu Wang, Wenlong Li, Xin Jin, Yiting Pan, Yunjie Sheng, Huaqiang Li, Luping Qin, Xiongyu Meng","doi":"10.3390/md23030131","DOIUrl":"10.3390/md23030131","url":null,"abstract":"<p><p>Diterpenoids from marine-derived organisms represent a prolific source of secondary metabolites, characterized by their exceptionally promising chemical structures and pronounced pharmacological properties. In recent years, marine diterpenoids have garnered considerable attention and are regarded as a prominent area of scientific research. As a vital class of metabolites, diterpenoids show diverse biological activities, encompassing antibacterial, antifungal, antiviral, anti-inflammatory, inhibitory, and cytotoxic activities, among others. With the rapid advancement of equipment and identified technology, there has been a tremendous surge in the discovery rate of novel diterpenoid skeletons and bioactivities derived from marine fungi over the past decade. The present review compiles the reported diterpenoids from marine fungal sources mainly generated from January 2000 to December 2024. In this paper, 515 diterpenoids from marine organisms are summarized. Among them, a total of 281 structures from various fungal species are included, comprising 55 from sediment, 39 from marine animals (predominantly invertebrates, including 17 from coral and 22 from sponges), and 53 from marine plants (including 34 from algae and 19 from mangrove). Diverse biological activities are exhibited in 244 compounds, and among these, 112 compounds showed great anti-tumor activity (45.90%) and 110 metabolites showed remarkable cytotoxicity (45.08%). Furthermore, these compounds displayed a range of diverse bioactivities, including potent anti-oxidant activity (2.87%), promising anti-inflammatory activity (1.64%), great anti-bacterial activity (1.64%), notable anti-thrombotic activity (1.23%), etc. Moreover, the diterpenoids' structural characterization and biological activities are additionally elaborated upon. The present critical summary provides a comprehensive overview of the reported knowledge regarding diterpenoids derived from marine fungi, invertebrates, and aquatic plants. The systematic review presented herein offers medical researchers an extensive range of promising lead compounds for the development of marine drugs, thereby furnishing novel and valuable pharmaceutical agents.</p>","PeriodicalId":18222,"journal":{"name":"Marine Drugs","volume":"23 3","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11943766/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143710274","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}