Next Article in Journal
The Effect of Phosphorus Concentration on the Co-Production of Fucoxanthin and Fatty Acids in Conticribra weissflogii
Previous Article in Journal
Isolation and Characterization of Bioactive Compounds from Saccharomonospora sp. CMS18 and Their Antifungal Properties
Previous Article in Special Issue
Protective Effect of Marine Peptide from Netunea arthritica cumingii Against Gentamicin-Induced Hair Cell Damage in Zebrafish
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances of the Zebrafish Model in the Discovery of Marine Bioactive Molecules

1
Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
2
Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensor of Shandong Province, Jinan 250103, China
*
Authors to whom correspondence should be addressed.
Mar. Drugs 2024, 22(12), 540; https://doi.org/10.3390/md22120540
Submission received: 28 October 2024 / Revised: 27 November 2024 / Accepted: 27 November 2024 / Published: 30 November 2024
(This article belongs to the Special Issue Zebrafish Models in Marine Drug Discovery)

Abstract

:
Marine natural products are increasingly utilized in nutrition, cosmetics, and medicine, garnering significant attention from researchers globally. With the expansion of marine resource exploration in recent years, the demand for marine natural products has risen, necessitating rapid and cost-effective activity evaluations using model organisms. Zebrafish, a valuable vertebrate model, has become an efficient tool for screening and identifying safe, active molecules from marine natural products. This review, based on nearly 10 years of literature, summarizes the current status and progress of zebrafish models in evaluating marine natural product bioactivity. It also highlights their potential in exploring marine resources with health benefits, offering a reference for the future development and utilization of marine biological resources.

1. Introduction

Covering over 70% of the Earth’s surface, the ocean is the cradle of life and the largest known habitat. The marine biosphere hosts a vast diversity of species and immense biomass, comprising over 80% of the Earth’s total biomass, making it a significant reservoir of natural resources [1,2]. Compared to terrestrial environments, marine conditions—such as high salinity, pressure, oligotrophy, darkness, and extreme temperatures—have driven marine organisms to develop unique metabolic pathways and defense mechanisms. These adaptations enable the production of biologically active substances with complex and diverse structures, offering valuable opportunities to discover novel marine-derived compounds with bioactive potential [3,4]. In recent years, numerous marine compounds with antitumor, anti-inflammatory, antioxidant, and cardiovascular protective properties have been identified. These compounds hold significant promise for use in nutrition, cosmetics, pharmaceuticals, and other biotechnology sectors closely linked to human health [5]. Consequently, marine natural products have drawn extensive attention from researchers worldwide. In medicine, marine natural products are a major source of modern drugs [6]. Currently, 20 marine-derived drugs have received market approval, with dozens more in clinical trials, playing pivotal roles in treating cardiovascular diseases, cancer, and infections [7].
Bioactivity screening is a crucial step in drug discovery. Traditional in vitro activity screening, based on cells and enzymes, has evolved to offer high-throughput, rapid, sensitive, and cost-effective evaluations, thanks to advances in automation and instrumentation [8]. However, in vitro screening has its limitations. For example, in vitro cultured cells cannot replicate the extracellular microenvironment or disease progression. Many specialized cell types and organs cannot be cultured in vitro [9]. In contrast, in vivo bioactivity assays based on animal models provide more biologically relevant insights. Bioactive natural products identified in vivo are more likely to show activity in mammalian models [10]. Common animal models include invertebrates (e.g., nematode, fruit fly), non-mammalian vertebrates (e.g., zebrafish), and mammals (e.g., mouse). Among these, zebrafish (Danio rerio) has emerged as an excellent model, bridging flies/nematodes and mice/humans for studying embryonic development and gene function [8,11].
First introduced as an experimental model in the 1950s [12,13], zebrafish embryos were later used to study genetics, bringing zebrafish to the forefront of experimental biology [14,15]. The use of zebrafish for chemical screening in microplates began in 2000 [16]. Since then, zebrafish have become a powerful model for rapidly analyzing gene function and bioactivity. Several reviews have discussed zebrafish’s role in screening for antitumor, cardiovascular, and neuroprotective natural products, as well as safety evaluations [8,17,18,19]. However, there remains a gap in reviews specifically addressing the use of zebrafish models in evaluating marine natural product bioactivity. This article aims to fill this gap by summarizing the application of zebrafish in screening marine bioactive natural products. It covers zebrafish biology and key studies involving zebrafish in discovering marine bioactive molecules for nearly 10 years. The bioactivities discussed in this article include cardioprotection, proangiogenesis, antiangiogenesis, antithrombosis, anticancer, antidiabetes, anti-obesity, antioxidation, and anti-inflammation (Figure 1). This review seeks to raise awareness of zebrafish models, expand their application, and promote the development of marine natural products.

2. Zebrafish as a Model Species

Zebrafish are small tropical freshwater fish belonging to the genus Danio in the family Cyprinidae. They originate from South Asia, including countries such as Bangladesh and India. Adult zebrafish are 3–5 cm in length, with a slender, spindle-shaped body featuring blue and silver stripes on their surface [20,21]. Zebrafish are easy to breed due to their gentle temperament and tolerance to various environmental conditions. Optimal laboratory breeding conditions include a temperature of 28 °C, pH levels of 7–8, a 14:10 h light–dark cycle, and salinity of 0.25–0.6 ppm [17,22]. Zebrafish embryos are small, measuring 1–5 mm, and develop rapidly, reaching adulthood within approximately three months. The reproductive cycle is about one week, and a female can lay 200–300 eggs in one spawning [9].
Zebrafish are ideal for studying tissue and organ development due to their rapid development and short sexual maturity cycle. They are highly reproductive, providing an abundant supply of embryos for experiments. Embryos and larvae are small, require minimal amounts of test samples, and are easy to handle in microplates [23]. The embryos are optically transparent, allowing in vivo observation of organ and tissue development without damage. The simple administration of test samples, which can be diluted in sterile water or dimethyl sulfoxide, enables quick absorption through the zebrafish’s skin and gills [8,24]. Zebrafish can generate large numbers of synchronously developing transparent embryos, ensuring that all relevant tissues and organs form within a few days post-fertilization. This minimizes the sample amount needed for in vivo experiments at microgram levels, enabling high-throughput phenotypic screening on microplates, which is essential for discovering bioactive natural products [25,26]. The zebrafish model offers high reproducibility, providing reliable statistical data for validation and allowing rapid in vivo bioactivity analysis at microgram levels, even with limited sample sizes [8]. Determining biological activity in zebrafish offers advantages such as high sensitivity, stable internal testing, and the ability to detect chemicals with similar modes of action or sites of activity [27].
Zebrafish share significant physiological and genetic similarities with mammals. They have more than 70% genome homology with humans and up to 82% homology with human disease-related genes. Zebrafish also possess genes associated with the cell cycle, growth, differentiation, tissue function, tumorigenesis, and suppression [28]. Compared to fruit flies and nematodes, zebrafish represent a more complete vertebrate system, with physiological and pharmacological responses similar to those of humans [29]. These characteristics make zebrafish highly suitable for identifying drugs and bioactive natural products with therapeutic potential [30]. Additionally, zebrafish provide economic and experimental advantages over mammalian models, reducing the number of mammals used in research, cutting maintenance costs, shortening experimental cycles, and facilitating high-throughput pharmacological screening—all in adherence to the 3R principles (Replacement, Reduction, and Refinement) [22,31,32]. Therefore, zebrafish offer distinct advantages over fruit flies, nematodes, and mammals in biological and biomedical research (Table 1). Their whole biological complexity combined with high-throughput screening capabilities makes zebrafish an ideal model for screening marine bioactive natural products, which have low production and are difficult to obtain.
Since the mid-1990s, large-scale genetic screenings have produced hundreds of zebrafish mutant lines with various disease-related phenotypes [10]. Following the completion of zebrafish genome sequencing, research using zebrafish models has progressed rapidly. Techniques such as gene editing and transgenesis have facilitated the construction of various transgenic zebrafish lines with specific fluorescent protein expression in cells, tissues, and organs. These models have been successfully applied to pathological studies and drug screening [9,33,34,35,36]. For example, transgenic zebrafish Tg(fli1: EGFP) are used for screening vascular promoters and inhibitors, Tg(CD41: EGFP) for antithrombotic drugs and Tg(cmlc2: GFP) for cardioprotective drugs. Casper mutant zebrafish are commonly employed in cancer and stem cell research [37,38,39,40,41]. Zebrafish models also play a vital role in toxicology research, elucidating the functions of specific genes and their interactions with signaling pathways. The Wellcome Trust Sanger Institute in the UK has established over 3000 mutant zebrafish lines related to human diseases through large-scale mutagenesis screening [42].

3. Zebrafish Models for Evaluating Marine Bioactive Natural Products

Choosing an appropriate high-throughput screening model is essential for identifying lead compounds in drug discovery. With innovations in zebrafish disease models and advancements in screening technologies, zebrafish have become increasingly valuable for screening marine bioactive natural products. Various zebrafish models have been employed to efficiently explore marine bioactive substances. Below, we highlight the progress of zebrafish models in the investigation of marine bioactive compounds (Table 2), particularly regarding cardiovascular diseases, cancers, metabolic diseases, inflammation, and oxidative stress over the past decade.

3.1. Zebrafish Models Related to Cardiovascular Diseases

Cardiovascular diseases encompass various conditions affecting the heart and vascular system, with complex etiologies involving environmental and genetic factors. These diseases pose significant health risks, emphasizing the need for safe, effective cardiovascular drugs. Zebrafish exhibit strong similarities to humans in terms of cardiac action potentials, electrocardiograms, vascular structures, and genes governing heart development. The conservation of cardiovascular drug responses between zebrafish and humans underpins zebrafish’s role in cardiovascular drug discovery and toxicology research [43,44]. Zebrafish embryos’ transparency and rapid cardiovascular development enable non-invasive in vivo imaging of heart function. Structural and functional changes, including heart chambers, heartbeat, blood flow, and vasculature, are easily observed. Transgenic zebrafish expressing fluorescent proteins in endothelial, cardiac, or red blood cells are frequently used to study cardiovascular and hematopoietic system development [45]. Here, we review the use of zebrafish models to discover marine natural products with cardioprotective, antithrombotic, and vascular regulatory properties.

3.1.1. Evaluation of Cardioprotective Activity by Zebrafish Models

Cardiac function is crucial for diagnosing and treating cardiovascular diseases. The zebrafish heart, composed of the sinus venosus, atrium, ventricle, and bulbus arteriosus, is located in the anterior abdomen between the pleura and thorax [46]. By 48 h post-fertilization, zebrafish embryos have typically completed heart development and begin circulating blood through contraction and relaxation [47]. Therefore, zebrafish at this stage are commonly used to evaluate cardioprotective activity. Key indicators of cardiac function include heart morphology, heart rate, sinus venosus–bulbus arteriosus (SV–BA) distance, stroke volume, and ejection fraction [48]. Gene editing techniques have produced zebrafish strains expressing fluorescent proteins in the heart, such as Tg(cmlc2:EGFP), Tg(myl7:EGFP) and Tg(-5.1myl7:DsRed2-NLS) [48]. Using these strains, models of heart failure, arrhythmias, and other cardiac diseases have been established using drugs like verapamil, doxorubicin, terfenadine, and aconitine. These models facilitate the screening and identification of bioactive molecules with cardioprotective effects.
Currently, zebrafish cardioprotective models have been successfully established and applied for screening marine molecules with anti-heart failure and anti-arrhythmic effects. For example, using the AB or Tg(cmlc2:EGFP) zebrafish strain, our group developed cardioprotective models with verapamil and terfenadine. We identified phospholipids from shrimp heads, squid gonads, and viscera, such as phosphatidylcholine and phosphatidylethanolamine, which demonstrated anti-heart failure and anti-arrhythmic effects [49,50,51,52]. Additionally, a peptide PcShK3 from the zoantharian Palythoa caribaeorum exhibited significant cardioprotective activity at low concentrations [53]. Pott et al. discovered okadaic acid and calyculin A from sponges, using ILK-deficient msq mutant zebrafish embryos, which showed anti-heart failure activity [54]. Zebrafish-based heart failure and arrhythmia models provide a valuable platform for identifying cardioprotective marine molecules, supporting their further development and application.

3.1.2. Evaluation of Angiogenesis Regulatory Activity by Zebrafish Models

Angiogenesis, the formation of new capillary networks, involves endothelial cell proliferation and migration. In zebrafish, fluorescent reporter genes driven by vascular-specific promoters, such as flk1:EGFP or fli1:EGFP, allow direct visualization of angiogenesis [55]. Angiogenesis primarily occurs via sprouting, regulated by vascular endothelial growth factor (VEGF) [56]. VEGF-A, secreted by somatic cells, is crucial for inducing intersegmental vessel growth in the dorsal aorta and serves as a target for angiogenic therapies [57,58]. Angiogenesis, regulated by proangiogenic and inhibitory factors, is critical for maintaining vascular balance. Disruption of this balance may result in excessive or reduced angiogenesis, contributing to various diseases [56].
Inadequate angiogenesis can lead to ischemic cardiovascular diseases, such as myocardial infarction, angina pectoris, and coronary heart disease [59]. Developing angiogenesis-promoting drugs may aid in treating these conditions, and marine natural products offer potential sources. Zebrafish models have identified several marine compounds with proangiogenic activity. Using Tg(fli-1:EGFP) or Tg(vegfr2:GFP) zebrafish lines, our team established an intersegmental vessel defect model with PTK787, discovering many compounds with proangiogenic activity like dinotoamide J [60], communesin I, fumiquinazoline Q, and protuboxepin E [61], N-(2-hydroxyphenyl)-acetamide, ethyl formyltyrosinate [62], chaetofanixins A–E [63], bialorastin C [64], sterigmatocystin A [65], agelanemoechine [66], lemnardosinanes A [67], marchaetoglobin B and C [68], chevalinulins A and B [69], clavukoellian K [70], chaetoviridin L, chaetomugilin A, and chaephilone D [71]. Additionally, marine compounds like fucoidan LMWF [72], ZoaNPY [73], Cyclotripeptide X-13 [59], and Pestaphilone J [74], as summarized in Table 2.
Diseases linked to excessive angiogenesis, including tumors, retinopathies, rheumatoid arthritis, and moyamoya disease, require anti-angiogenic therapies [75]. Currently, anti-angiogenic research primarily focuses on inhibiting tumor angiogenesis, which plays a pivotal role in tumor growth, invasion, and metastasis and is recognized as one of cancer’s hallmarks [76]. Several anti-angiogenic drugs have been approved for cancer treatment. Numerous anti-angiogenic compounds have been identified from marine sources using zebrafish models (Table 1), including pyrrolidinedione AD0157 [76], catunaregin [77], bis(2,3-dibromo-4,5-dihydroxybenzyl) ether (BDDE) [78], stellettin B [79], Ishophloroglucin A [80], polypeptide CS5931 [81], Diphlorethohydroxycarmalol [82], protein ASP-3 [83], polysaccharide SPS [84], quinadoline B [85], phloroglucinol and dieckol [86], Sinularin [87], capnellene GB9 [88], Toluquinol [89], Solomonamide A [90], Somocystinamide A [91], Fucoidan [92], dihydroaustrasulfone alcohol WA-25 [93], bis(2,3,6-tribromo-4,5-dihydroxybenzyl) ether (BTDE) [94], asperhiratide [95], pyrrole-pyridinimidazole derivative 8a [96], penisterine C and D [97], monacolin X [98], Dolastatin 15 [99], and murrangatin [100].

3.1.3. Evaluation of Antithrombotic Activity by Zebrafish Models

Thrombosis is a serious cardiovascular condition posing a significant threat to human life. Genes related to coagulation, anticoagulation, and platelet signaling in zebrafish show high homology with those in humans. Studies have shown that internal and external coagulation factors, as well as multiple platelet surface receptors found in human blood and platelets, also exist in zebrafish [101]. Zebrafish possess cellular and humoral hemostatic mechanisms similar to those in humans, making them an excellent model for studying thrombosis and antithrombotic drugs. Zebrafish exhibit similar responses to commonly used clinical anticoagulants and antiplatelet drugs, thus mimicking human hemostasis and thrombosis [102,103]. The transparency of zebrafish embryos allows dynamic observation of arterial or venous thrombosis formation under a microscope, and zebrafish thrombus models are stable and reproducible, making them a valuable tool for antithrombotic research.
Blood clot formation can result in myocardial infarction, ischemia, and stroke, which are major global causes of death. Developing effective, safe therapies for thrombotic diseases is a significant challenge [104]. Zebrafish thrombosis models can be induced by phenylhydrazine (PHZ) [105], ferric chloride [106,107], arachidonic acid [108], microinjection, or direct immersion. These models have been instrumental in identifying safe, effective antithrombotic natural products. Antithrombotic efficacy is typically assessed by staining wild-type zebrafish larvae with o-dianisidine and evaluating the red blood cell staining intensity in the heart or observing venous thrombosis morphology under a fluorescence microscope. Additionally, transgenic zebrafish strains such as Tg(CD41:EGFP) and Tg(LCR:EGFP), which express green fluorescent protein in platelets and red blood cells, offer models for dynamically evaluating antithrombotic activity [109].
Various marine natural products with antithrombotic activity have been identified (Table 2). For example, using the arachidonic acid-induced thrombosis model of the AB zebrafish strain, our group discovered that 4-hydroxyphenylacetic acid [110], sarcoeleganolide I [111], sarcoelegan C [112], and sarcocinerenolides C and H [113], derived from marine fungi and soft corals, as well as phospholipids from shrimp heads, squid viscera, and gonads [49,50,51,52], exhibit antithrombotic effects. Additionally, Yang et al. identified a pyruvylated and sulfated galactan (PSG) with antithrombotic activity from the green alga Dictyosphaeria cavernosa using the phenylhydrazine-induced thrombosis model in Tg(gata1:dSRed) and Tg(CD41:EGFP) zebrafish strains [114].

3.2. Zebrafish Models Related to Cancer

Zebrafish have become essential animal models for human tumor research, aiding in the preclinical development of anticancer drugs. Various zebrafish models for cancer research have been developed, categorized into induced, transgenic, and implanted models [17,115]. Among these, transgenic and implanted zebrafish tumor models have been applied to marine natural product research.

3.2.1. Evaluation of Anticancer Activity by Transgenic Zebrafish Models

Transgenic zebrafish models are developed through gene knockdown or gene editing in the whole organism or specific organs [116]. Zebrafish share significant homology with human cancer-related genes, making gene manipulation feasible. These models have rapidly become valuable tools for cancer research [17,117]. One model involves inducing tumors by regulating the abnormal expression of oncogenes or tumor suppressor genes. For instance, a T-lymphocyte leukemia model is created by overexpressing the mouse oncogene c-myc under the zebrafish lymphocyte gene promoter rag2 [118]. Additionally, by knocking out the tumor suppressor gene Rb1 in a mitf-BRaf and p53 −/− background, highly aggressive melanoma cells are induced, creating an immune-compatible melanoma zebrafish model [119]. Compounds like Oligo-Fucoidan [120] and Terphenyllin Derivative CHNQD-00824 [121] have shown antitumor activity using this model. Though many such models are available, few have been used to evaluate the anticancer potential of marine natural products.
Another important model for cancer research involves inhibiting angiogenesis, a process critical for tumor growth and metastasis. As effective angiogenesis inhibitors are key in cancer treatment [122], these zebrafish models of antiangiogenesis activity are commonly used to screen for anticancer compounds from marine sources. Reviews have summarized transgenic zebrafish models used in cancer research, highlighting key developments, advantages, and limitations [117,122,123,124].

3.2.2. Evaluation of Anticancer Activity by Xenograft Zebrafish Models

Implanted zebrafish tumor models, primarily xenograft models, involve implanting fluorescent protein-labeled tumor cells or tissues into zebrafish embryos or adults. This allows real-time observation of cancer cell proliferation and metastasis and is a widely used method with a high success rate [116]. The optimal time for tumor cell implantation is within 48 h post-fertilization. Implantation sites include the yolk sac, perivitelline space, and duct of Cuvier, as well as the abdominal cavity in adult zebrafish [115,125]. Since the first zebrafish xenograft model was introduced in 2005, the technique has matured and is widely used in different cancer types [123]. These models use commercial human cancer cell lines or primary tumor cells from cancers such as melanoma, breast, colorectal, pancreatic, ovarian, renal, lung, and oral cancers [126]. Researchers can control tumor cell implantation in different zebrafish locations to meet specific research goals. Numerous marine antitumor molecules have been discovered using these models (Table 2), including rakicidin B and B1 [127], rhopaloic acid A [128,129], tilapia piscidin 4 [130], crambescidine-816 [131], holothurian glycosaminoglycan [132], saringosterol acetate [133], cyclo(l-Pro-l-Leu), cyclo(l-Pro-l-Val), cyclo(l-Pro-l-Phe), cyclo(l-Pro-l-Tyr) [134], intestinal peptide SCIP [135], anticancer peptides MP06 [136], isofistularin-3 [137], and trabectedin [138].

3.3. Zebrafish Models Related to Metabolic Disorder

Metabolic disorders like diabetes and obesity, along with related conditions like hyperglycemia, hyperlipidemia, hypertension, and fatty liver disease, are emerging as major public health concerns [139,140,141]. Zebrafish models have proven useful for studying these conditions and exploring treatments. Their advantages make them an ideal model for screening marine natural products that may prevent or treat metabolic disorders [142,143].

3.3.1. Evaluation of Antidiabetic Activity by Zebrafish Models

Diabetes is a condition characterized by chronic hyperglycemia [144]. The morphogenesis and basic cellular structure of the zebrafish pancreas shares similarities with mammals, consisting of an exocrine gland (ductal, acinar, and centroacinar cells) and an endocrine gland (α-cells, β-cells, δ-cells, ε-cells, and pp-cells) [145]. Hormones like glucagon, insulin, and somatostatin regulate glucose levels [146,147]. The development and signaling pathways involved in the zebrafish endocrine pancreas are highly homologous to mammals, and the exocrine gland is morphologically and ultrastructurally conserved, making zebrafish a suitable model for diabetes research [142]. Normal blood glucose levels in zebrafish (50–75 mg/dL) are comparable to humans (70–120 mg/dL) [148].
Diabetes is mainly categorized into type 1 and type 2. Type 1 diabetes is a chronic autoimmune destruction characterized by the loss of insulin-producing β-cells in the pancreas, leading to insulin deficiency, and often occurs in children [149]. Type 2 diabetes is primarily due to acquired insulin resistance, which usually occurs along with decreased insulin secretion, accounts for 90% of all diabetes cases, and is often associated with obesity [150,151]. Diabetic zebrafish models are created using chemical induction, glucose immersion, transgenic techniques, or gene knockout, resulting in hyperglycemia and insulin resistance [152,153]. Several antidiabetic components have been identified from marine natural products using zebrafish models (Table 2), including brasilterpenes A and C [154], penipyrol C [155], Polysiphonia japonica extracts and 5-bromoprotocatechualdehyde [156,157], Con-Ins peptides [158], aspterric acid [159], Antarctic krill enzymatic hydrolysates [160], and dieckol [161].

3.3.2. Evaluation of Anti-Obesity Activity by Zebrafish Models

Obesity is a complex metabolic disorder characterized by excessive lipid accumulation in the body and significant increase in free fatty acids and triglycerides in the serum and often caused by high-fat, high-sugar diets and lack of exercise [17]. It increases the risk of chronic diseases such as diabetes, cardiovascular disease, and cancer [141]. The lipid metabolism networks and the hypothalamic circuits regulating energy balance in zebrafish are highly conserved with humans [162,163,164]. Zebrafish store excess nutrients in white adipocytes as lipid droplets, which can be visualized using neutral dyes like Oil Red O or Nile Red, facilitating the study of hypolipidemic effects [165,166]. The similarities in lipid storage and metabolism, along with zebrafish’s advantages (short life cycle, ease of use, and low drug dosage), make them ideal for obesity research and anti-obesity drug screening.
Obese zebrafish models can be generated via diet-induced obesity or transgenic technology. Diet-induced models are primarily used for lipid-lowering screenings, while genetically engineered models provide insight into obesity mechanisms [167]. Using these models, several marine compounds with anti-obesity and lipid-lowering effects have been discovered (Table 2). For example, Ralph Urbatzka’s team conducted a zebrafish Nile red fat metabolism assay by a diet-induced obese zebrafish model and found that extracts from seagrass, blue-green algae, and marine actinomycetes, along with compounds like 13-2-hydroxypheophytine, vitamin K1-analog (OH-PhQ), citreorosein, questinol, (1′Z)-2-(1′,5′-dimethylhexa-1′,4′-dieny1)-5-methylbenzene-1,4-diol, 6-(3-hydroxy-6-methyl-1,5-heptadien-2-yl)-3-methylbenzene-1,4-diol, 4-hydroxy-3,7-dimethyl-7-(3-methylbut-2-en-1-yl)benzofuran-15-one, 1,8-epoxy-1(6),2,4,7,10-bisaborapentaen-4-ol, and 9-(3,3-dimethyloxiran-2-yl)-1,7-dimethyl-7-chromen-4-ol, exhibited anti-obesity activities [168,169,170,171,172,173,174,175,176,177]. Additionally, studies have shown that red seaweed Palmaria mollis [178], polysaccharide SS3-N1 [179], fucoxanthin [180], glycosaminoglycans [181], and saringosterol acetate [182] exhibit promising anti-obesity effects.

3.4. Zebrafish Models Related to Inflammation

Inflammation is a series of defensive physiological responses to internal and external stimuli, involving multiple cytokines that regulate the inflammatory system’s balance [183]. Excessive or prolonged inflammation can cause the immune system to damage normal tissues, impair functions, and trigger pathological reactions [184]. Diseases like cardiovascular disease, cancer, diabetes, and Alzheimer’s are linked to inflammation [185,186]. The zebrafish have highly conserved natural and acquired immune systems, and its immune cell types, immune signal transduction and functions are similar to those of mammals [187]. Zebrafish natural immune systems can quickly respond to infections and tissue damage, easily causing inflammatory reactions, making them ideal for studying inflammation and wound repair [188]. The development of transgenic zebrafish strains with fluorescent immune cell markers, along with the transparency of zebrafish embryos, allows for real-time tracking of immune responses [189].
The triggering factors of inflammation include mechanical damage, chemical induction, bacterial and viral infections, etc. At present, zebrafish experiments mainly use three methods to simulate inflammation of the immune system: local inflammation induced by tail cutting, systemic inflammation induced by lipopolysaccharide, and acute inflammation induced by copper sulfate. Several transgenic zebrafish strains have been developed, such as Tg (lysC: EGFP), Tg (lysC: DsRED2), and Tg (coro1a: EGFP), to label neutrophils and macrophages [186]. The zebrafish inflammation model is widely used to discover marine bioactive molecules (Table 2). Our team used Tg (zlyz: EGFP) zebrafish to identify anti-inflammatory compounds, including peptide LLTRAGL [190], Septosone A [191], Somallactam A [192], Dysidinoid B [193], Sarcoelegan variants [112], Altechromone A [194], Purpurols A and B [195], and protein hydrolysate AJH-1 [196]. Other teams have identified molecules like apo-9′-fucoxanthinone [197], 5-hydroxypalisadin B [198], fucoidan SFF-PS-F5 [199], fucoidan LJSF4 [200], fucose containing sulfated polysaccharides FCSP [201], 24-Methylcholesta-5, 22-Diene-3β-ol [202], sulfated polysaccharides CFCE-PS [203], sulfated polysaccharides SFPS [204], sulfated Galactofucan LJNF3 [205], maxol and dieckol [206], enzymatic peptide SEP [207], and Micrometam C [208].

3.5. Zebrafish Models Related to Oxidative Stress

Oxidative stress occurs when the body’s oxidation or antioxidant systems become imbalanced, leading to excessive production of reactive oxygen species (ROS) [209]. When the original dynamic balance of redox is disrupted, a large amount of oxidative intermediate ROS are generated and cause cell structural changes and genetic damage [210]. Prolonged oxidative stress can result in pathological conditions, including cardiovascular disease, inflammation, cancer, and metabolic disorders [211]. Zebrafish genes involved in oxidative processes are highly conserved, and many proteins share functions with their mammalian counterparts [212]. Zebrafish have become a valuable model organism for studying antioxidant mechanisms and screening marine-derived antioxidant compounds in vivo.
The construction of zebrafish oxidative stress models mainly involves two methods: the physical hypoxia method and chemical drug intervention. The physical hypoxia method uses nitrogen perfusion and Gaspak anaerobic techniques, while the chemical drug intervention method utilizes compounds such as AAPH, sodium sulfite, and H2O2 to induce oxidative stress [209]. After oxidative induction, indicators like ROS, nitric oxide, antioxidant enzyme activity, malondialdehyde, heart rate, and mortality rate are measured to evaluate the antioxidant capacity of natural products. Zebrafish oxidative stress models have been widely applied in discovering marine bioactive molecules (Table 2). For instance, using metronidazole-induced Tg (krt4: NTR hKikGR)cy17 zebrafish, our team identified peptides YSQLENEFDR and YIAEDEAR with antioxidant activity from sea snails [213], and Frondoplysin A from the marine sponge Dysidia fructosa [214]. Other antioxidant components, such as peptide AFFP [215], seahorse peptide SHP [216], phenoglucinol, eckol, dieckol, and trihalomethaneol A [217], agaro-oligosaccharides AO [218], xyloketal B [219], (−)-loliolide [220], protein hydrolysate HPH [221], and fucoidan HFPS-F4 [222], have also been discovered.
Table 2. Application of zebrafish models in the bioactivity evaluation of marine natural products.
Table 2. Application of zebrafish models in the bioactivity evaluation of marine natural products.
Marine Nature ProductsSourceModelsMain Effects (Concentrations)References
PhospholipidsShrimp heads, squid gonads, and visceraAB or Tg(cmlc2: EGFP) zebrafish strainAnti-heart failure and anti-arrhythmic effects (25, 50, 100 μg/mL and 20, 40, 80 μg/mL)[49,50,51,52]
Peptide PcShK3zoantharian Palythoa caribaeorumTg(cmlc2: GFP) zebrafish strainCardio-protective activity (20 μM)[53]
Calyculin A and okadaic acidspongesILK-deficient msq mutant zebrafish embryosAnti-heart failure activity (100 and 0.15 μM)[54]
Dinotoamide JMarine-derived fungus Aspergillus austroafricanus Y32-2Tg (vegfr2: GFP) zebrafish modelProangiogenic activity (70, 120 μg/mL)[60]
Communesin I, Fumiquinazoline Q and Protuboxepin EMarine-derived fungus Penicillium expansum Y32Tg (vegfr2: GFP) zebrafish modelProangiogenic activity (20 and 50 and 100 μg/mL)[61]
N-(2-hydroxyphenyl)-acetamide, ethyl formyltyrosinateMarine-derived fungus Penicillium chrysogenum Y20-2Tg (FLI1: EGFP) zebrafish modelProangiogenic activity (25, 50, 100 μg/mL)[62]
Chaetofanixins A-EHadal trench-derived fungus Chaetomium globosum YP-106Tg (flk1: EGFP) zebrafish modelProangiogenic activity (20, 40, 80 μg/mL)[63]
Bialorastin CDeep-sea cold-seep-derived fungus Penicillium bialowiezense CS-283Tg(vegfr2: GFP) zebrafish modelProangiogenic activity (20, 40 μM)[64]
Sterigmatocystin ASponge-derived fungus Aspergillus versicolor (15XS43ZD-1)Tg (vegfr2: GFP) zebrafish modelpromoting angiogenesis activity (1.25 μM)[65]
AgelanemoechineSouth China sea sponge Agelas nemoechinataTg (vegfr2: GFP) zebrafish modelProangiogenic activity (5 μM)[66]
Lemnardosinanes ASoft coral Lemnalia sp.Tg (vegfr2: GFP) zebrafish modelProangiogenic activity (20 μM)[67]
Marchaetoglobin B and CMarine-sponge-associated fungus Chaetomium globosum 162105Tg (vegfr2: GFP) zebrafish modelProangiogenic activity (80 μM)[68]
Chevalinulins A and BDeep-sea cold-seep-derived fungus Aspergillus chevalieri CS-122Tg (vegfr2: GFP) zebrafish modelProangiogenic activity (40 and 80 μg/mL)[69]
Clavukoellian KMarine soft coral Lemnalia spTg (vegfr2: GFP) zebrafish modelProangiogenic activity (2.5 μM)[70]
Chaetoviridin L, chaetomugilin A, and chaephilone DHadal trench-derived fungus Chaetomium globosum YP-106Tg (flk1: EGFP) zebrafish modelProangiogenic activity (20, 40, 80 μg/mL)[71]
Fucoidan LMWFBrown algae
Saccharina japonica
High-glucose-induced zebrafish with blood vessel growth inhibitionPromotes subintestinal vessel formation in angiogenesis (100 μg/mL)[72]
Polypeptide ZoaNPYZoanthus sociatusTg (Fli1a: EGFP) zebrafish modelProangiogenic effects (100 pmol)[73]
Cyclotripeptide X-13Mangrove fungus Xylaria sp. (no. 2508)Tg (fli1: EGFP) zebrafish modelProangiogenic activity (10, 50, 100 μM)[59]
Pestaphilone JSea-mud-derived fungus Neopestalotiopsis sp. HN-1-6Tg (flk1:EGFP) zebrafish modelProangiogenic activity (40 μM)[74]
Pyrrolidinedione AD0157Marine fungiTg (fli1:EGFP)y1 zebrafish modelAnti-angiogenic activity (10 µM)[76]
CatunareginStem bark of Catunaregam spinosaTg (fli1: EGFP) zebrafish modelAnti-angiogenic activity (10, 50, 100 μM)[77]
Bis(2,3-dibromo-4,5-dihydroxybenzyl) ether (BDDE)Marine algae
Leathesia nana and
Rhodomela confervoides
Zebrafish with Alkaline phosphatase stainingAnti-angiogenic activity (6.25, 12.5, 25 μM)[78]
Stellettin BMarine-sponge Stelletta sp.Tg (fli1: EGFP)y1 transgenic zebrafishAnti-angiogenic activity (≥50 nM)[79]
Ishophloroglucin AIshige okamuraeTg (flk: EGFP) zebrafish with high glucose-induced angiogenesisAnti-angiogenic activity (0.015, 0.05, 0.15, 0.5 µM)[80]
Polypeptide CS5931Ciona savignyiZebrafish modelAnti-angiogenic
Activity (10, 20, 30 μg/mL)
[81]
Diphlorethohydroxycarmalolbrown alga
Ishige okamurae
Tg (flk: EGFP) zebrafish with high glucose-induced angiogenesisAnti-angiogenic
Activity (0.06, 0.2, 0.6, 2 μM)
[82]
Protein ASP-3Arca subcrenata LischkeTg (fli1: GFP) zebrafish modelAnti-angiogenic
Activity(18.8–150 μg/mL)
[83]
Polysaccharide SPSBrown seaweed Sargassum integerrimumTg (fli1a:EGFP)y1 zebrafish modelAnti-angiogenic
Activity (0, 1, 4 mg/mL)
[84]
Quinadoline Bmarine-derived fungus Aspergillus clavutus LZD32-24Tg (fli1a: EGFP) zebrafish modelAnti-angiogenic
Activity (2, 5, 10 μM)
[85]
Phloroglucinol and dieckolBrown alga Ecklonia cavaTg (flk: EGFP) zebrafish under high glucose conditionsAnti-angiogenic activity (0.24, 0.8, 2.4, 8 and
0.03, 0.1, 0.3, 1 μM)
[86]
SinularinSoft coral Sinularia flexibilisThe angiofluorescent zebrafishAnti-angiogenic activity (5 μM)[87]
Capnellene GB9Soft coral Capnella imbricataTg (fli: EGFP) zebrafish modelAnti-angiogenic activity (10 μM)[88]
ToluquinolMarine fungus Penicillium sp. HL-85-ALS5-
R004
Tg (fli1: EGFP)y1
zebrafish model
Anti-angiogenic activity (20 μM)[89]
Solomonamide AMarine algae Leathesia nanaZebrafish modelAnti-angiogenic activity (5, 10 μM)[90]
Somocystinamide AMarine microorganisms Lyngbya majusculaTg(fli1: EGFP) zebrafish modelAnti-angiogenic activity (80, 160, 300, 1.6, 3 μM)[91]
FucoidanFucus vesiculosusTg(fli1: EGFP) zebrafish modelAnti-angiogenic activity (300 μg/mL)[92]
Dihydroaustrasulfone alcohol WA-25 Soft coral
Cladiella australis
Tg(fli1: EGFP)y1 and Tg(kdrl: mCherryci5-fli1a: negfpy7) zebrafish modelAnti-angiogenic activity
(50 μM)
[93]
Bis(2,3,6-tribromo-4,5-dihydroxybenzyl)ether (BTDE)Marine red alga Symphyocladia latiusculaTg (flk1: EGFP) zebrafish modelAnti-angiogenic activity (2.5–10 μM)[94]
AsperhiratideSoft coral-derived fungus Aspergillus hiratsukae SCSIO 5Bn1003Tg (fli1: EGFP) zebrafish modelAnti-angiogenic activity[95]
Pyrrole-pyridinimidazole derivative 8aMarine sponge Agelas nakamuraiTg (flk1: EGFP) zebrafish modelAnti-angiogenic activity(100, 150 μM)[96]
Penisterine C and DMarine brown alga derived fungus, Penicillium
sumatraense SC29
Tg (fli1: EGFP) zebrafish modelAnti-angiogenic activity (10.2, 20.4 and 8.6, 17.2 μg/mL)[97]
Monacolin XFungi-NMK7 associated with marine
sponge
Tg (Kdr: EGFP)/ko1 zebrafish modelAnti-angiogenic activity
(0.5, 1 μM)
[98]
Dolastatin 15Marine cyanobacteriavhl+/hu2117 heterozygous parents carrying the fli1a:egfpy1 transgeneAnti-vascularization effect (6 μM)[99]
MurrangatinMarine
plant
Tg (fli1: EGFP) zebrafish modelAnti-angiogenic effects
(10, 50, 100 μM)
[100]
4-hydroxyphenylacetic acidMarine-derived fungus Emericellopsis maritima Y39–2Arachidonic Acid induced AB zebrafish modelAntithrombotic activity(164.3, 328.6 μM)[110]
Sarcoeleganolide ISoft coral Sarcophyton elegansArachidonic Acid induced AB zebrafish modelAntithrombotic activity (20 μM)[111]
Sarcoelegan CSoft coral Sarcophyton elegansArachidonic Acid induced AB zebrafish modelAntithrombotic activity (20 μM)[112]
sarcocinerenolides C and Hsoft coral Sarcophyton cinereumArachidonic Acid induced AB zebrafish modelAntithrombotic activity (20 μM)[113]
Pyruvylated and sulfated galactan (PSG)Green alga Dictyosphaeria cavernosaphenylhydrazine-induced thrombosis model of Tg(gata1: dSRed) and Tg(CD41: EGFP) zebrafish strainsAntithrombotic activity (100, 150 μg/mL)[114]
Oligo-FucoidanBrown seaweedAB, Tg (fabp10a: HBV-HBx-mCherry, myl7: EGFP), Tg (fabp10a: src, myl7: EGFP), Tg (fabp10a: HBV-HBx-mCherry, myl7: EGFP, p53−/+), Tg (fabp10a: src, myl7: EGFP, p53−/+)Prevents
radiation-induced fibrosis and secondary tumors (300 mg/kg)
[120]
Terphenyllin derivative CHNQD-00824Marine-derived compound libraryTg (fabp10: rtTA2s-M2; TRE2: EGFP krasG12V)inhibit DOX-induced liver-specific
enlargement (2.5, 5 μM)
[121]
Rakicidin B and B1Marine MicromonosporaZebrafish xenotransplantation model with HCT-8 tumor cellAntitumor (3, 10,30, 35, 40 ng/mL)[127]
Rhopaloic acid AMarine sponge Rhopaloeides sp.Zebrafish oral and Bladder cancer xenotransplantation modelAntitumor
effects against oral and Bladder cancer (0.03, 0.3 μg/mL)
[128,129]
Tilapia piscidin 4Nile tilapiaAB zebrafish bladder cancer modelAntitumor
effects against bladder cancer (0.3, 1, 3 μg/mL)
[130]
Crambescidine-816Marine sponge Crambe crambeZebrafish xenotransplantation model with colorectal carcinoma cellsantitumor activity against colorectal
carcinoma (1, 5, 10 μM)
[131]
Holothurian glycosaminoglycanSea cucumber
Holothuria leucospilota
AB/Tubingen zebrafish xenotransplantation model with B16F10 tumor cellAntitumor
Effects (1 μM)
[132]
Saringosterol acetateBrown alga Hizikia fusiformeTg(fli1: EGFP) zebrafish
hepatocellular carcinoma xenograft model
Suppress hepatocellular carcinoma growth and metastasis (2 or 5 μg/g)[133]
Cyclo (l-Pro-l-Leu), cyclo (l-Pro-l-Val), cyclo (l-Pro-l-Phe) and
cyclo (l-Pro-l-Tyr)
Exiguobacterium
acetylicum S01
zebrafish xenogroft model with HT-29 tumor cellsInhibit the tumor progression (50, 100, 150 μM)[134]
Intestinal peptide (SCIP)Sea cucumberAB zebrafish xenogroft model with MCF-7 tumor cellsInhibits the proliferation of MCF-7 tumor cells (27.8, 83.3, 250 μg/mL)[135]
Peptides MP06Green sea algae Bryopsis plumosaTg(kdrl: GFP) zebrafish xenogroft model with A549 cellsReduce metastatic dissemination (1, 2, 4, 10 μM)[136]
Isofistularin-3Marine
sponge Aplysina aerophoba
zebrafish xenogroft model with VampiroPC3 or Vampiro-SH-SY5Y cellsAntiproliferative activity (5, 10, 25, 50 μM)[137]
TrabectedinMarine derived Ecteinascidia turbinatazebrafish xenogroft model with NCI-H295R, MUC-1, and TVBF-7 cellsReduce ACC cell xenograft area and metastasis formation (15 nM)[138]
Brasilterpenes A and CDeep Sea-Derived Fungus
Paraconiothyrium brasiliense HDN15-135
Tg(-1.2ins:htBidTEON; LR) zebrafish modelHypoglycemic activity (0, 1, 10, 50, 200 μM)[154]
Penipyrol CMangrove derived fungus Penicillium sp. HDN-11-131Tg(-1.2ins: H2BmCherry) and Tg (-1.2ins: H2BmCherry) zebrafish modelAnti-diabetes (10 μM)[155]
Extracts of Polysiphonia japonica and 5-BromoprotocatechualdehydePolysiphonia japonicaTg(ins: EGFP) zebrafish modelProtects against palmitate-induced β-cell dysfunction (10 and 50 μM)[156,157]
Con-Ins G1, Con-Ins G3, Con-Ins T1A, Con-Ins T2, Con-Ins K1, Con-Ins K2Conus geographus, C.tulipa, C.kinoshitaiStreptozotocin-induced model of diabetes in zebrafishReduce blood glucose (65 ng/g)[158]
Aspterric acidMangrove sediment-derived
fungus Penicillium polonicum H175
Tg (Ins: htBidTEON; LR) zebrafish modelHypoglycaemic effect (10 μmol/L)[159]
Antarctic krill enzymatic hydrolysates AKEHAntarctic krillDiet-induced diabetic zebrafish modelHypoglycaemic effect (1.35,2.70, 5.40 g/L)[160]
DieckolEcklonia cavaAlloxan-induced diabetic zebrafish modelAnti-diabetes activity (1 µg/g body weight)[161]
Palmaria mollisThe red seaweed Palmaria mollisDiet-induced obese zebrafish modelAnti-obesity effects
(PM dose of 2.5% (w/w))
[178]
Citreorosein and questinolMarine sponge-associated fungus Talaromyces stipitatus KUFA 0207.Diet-induced obese zebrafish modelAnti-obesity effects
(5 μM)
[176]
(1′Z)-2-(1′,5′-dimethylhexa-1′,4′-dieny1)-5-methylbenzene-1,4-diol, 6-(3-hydroxy-6-methyl-1,5-heptadien-2-yl)-3-methylbenzene-1,4-diol, 4-hydroxy-3,7-dimethyl-7-(3-methylbut-2-en-1-yl)benzofuran-15-one, 1,8-epoxy-1(6),2,4,7,10-bisaborapentaen-4-ol, 9-(3,3-dimethyloxiran-2-yl)-1,7-dimethyl-7-chromen-4-olMarine
sponge Myrmekioderma sp.
Diet-induced obese zebrafish modelLipid-reducing activity (10 µM)[177]
Polysaccharide SS3-N1Suaeda salsa L. in coastal saline-alkali areasEgg yolk powder-induced hyperlipidemic zebrafish modelHypolipidemic activity (100 μg/mL)[179]
FucoxanthinMarine brown algaeEgg yolk powder-induced hyperlipidemic zebrafish modelInhibits lipid accumulation (3.125, 6.25, 12.5 μM)[180]
GlycosaminoglycansOstrea rivularisHyperlipidemic zebrafishHypolipidemic effect
(fed with 125, 250, 500 mg/(kg·day))
[181]
Saringosterol acetateSargassum fusiformeDiet-induced obese adult male zebrafishAnti-obesity
Activity (2.5% (w/w))
[182]
Extracts of cyanobacteriacyanobacteriaDiet-induced obese zebrafishLipid Reducing Activity (10 μg/mL) [168,169]
The Extracts of seagrass Halophila stipulaceaSeagrass Halophila stipulaceaDiet-induced obese zebrafishLipid Reducing Activity (2, 6 μg/mL)[170]
fractions of cyanobacteriacyanobacterial libraryDiet-induced obese zebrafishRepress intestinal lipid absorption (10 μg/mL)[171]
Exometabolome from CyanobacteriaCyanobacteriaDiet-induced obese zebrafishLipid-Reducing Activity (25 µg/mL)[172]
Chlorophyll derivative
13-2-hydroxypheophytine
CyanobacteriaDiet-induced obese zebrafishReduce neutral lipid reserves (7.5 μg/mL)[173]
Vitamin K1-analog (OH-PhQ)Cyanobacterium Tychonema sp. LEGE 07196Diet-induced obese zebrafishLipid reducing activity (10 μg/mL)[174]
The Extracts of Microbacterium foliorum #91-29 and #91-40Microbacterium foliorumDiet-induced obese zebrafishLipid reducing activity (10 μg/mL)[175]
Peptide LLTRAGLRapana venosa2,4,6-trinitrobenzene sulfonic acid-induced Tg(zlyz: EGFP) zebrafish modelProtective effect against inflammatory bowel disease (20, 40, 80 μg/mL)[190]
Septosone AMarine Sponge Dysidea
septosa
CuSO4-induced Tg(zlyz: EGFP) zebrafish modelAnti-inflammatory activity (2.5, 5, 10 μM)[191]
Somalactam AStreptomyces somaliensis 1107LPS-induced Tg(zlyz: EGFP) zebrafish modelAnti-inflammatory effect (10 μM)[192]
Dysidinoid BSponge
Dysidea septosa
CuSO4-induced Tg(zlyz: EGFP) zebrafish modelAnti-inflammatory activity (20, 40, 80 μM)[193]
Sarcoelegan A, (±)-sarcoelegan D, sarcoelegan E, (+)-sarcoelegan F, and (+)-sarcoelegan HSoft coral Sarcophyton elegansCuSO4-induced Tg(zlyz: EGFP) zebrafishAnti-inflammatory activity (20 μM)[112]
Altechromone AMarine-derived fungus Penicillium Chrysogenum (XY-14-0-4)CuSO4-, tail-cutting-, and LPS-induced Tg(zlyz: EGFP) zebrafish and TNBS-induced zebrafish modelAnti-inflammatory activity (12.5, 25, 50 μg/mL)[194]
Purpurols A and BSponge Pseudoceratina purpureaCuSO4-induced Tg(zlyz: EGFP) zebrafishAnti-inflammatory activity (40 µM)[195]
Protein hydrolysate AJH-1sea cucumber Apostichopus japonicusCuSO4-induced Tg(zlyz: EGFP) zebrafishAnti-inflammatory activity (100, 250, 500 µg/mL)[196]
Apo-9′-fucoxanthinoneSargassum muticumLPS-induced zebrafish modelAnti-inflammatory activity (25, 50, 100 μg/mL)[197]
5-Hydroxypalisadin BRed seaweed
Laurencia snackeyi
LPS-induced and tail-cutting-zebrafish modelAnti-inflammatory activity (0.25, 0.5, 1 μg/mL)[198]
Fucoidan (SFF-PS-F5)Fermented Sargassum fusiformeLPS-induced zebrafish modelAnti-inflammatory activity (25, 50, 100 μg/mL)[199]
Fucoidan LJSF4Saccharina japonicaLPS-induced zebrafish modelAnti-inflammatory effect (12.5–50 μg/mL)[200]
Fucose containing sulfated polysaccharides (FCSP)Turbinaria ornata from the MaldivesLPS-induced zebrafish modelAnti-inflammatory effect (12.5, 25, 50 μg/mL)[201]
24-Methylcholesta-5(6), 22-Diene-3β-olMarine Diatom Phaeodactylum tricornutumLPS-induced zebrafish modelAnti-inflammatory effect (12.5, 25, 50 μg/mL)[202]
Sulfated polysaccharides (CFCE-PS)Green seaweed Codium fragileLPS-induced zebrafish modelAnti-inflammatory effect (25, 50, 100 μg/mL)[203]
Sulfated polysaccharides (SFPS)Brown Seaweed Sargassum fulvellumLPS-induced zebrafish modelAnti-inflammatory effect (25, 50, 100 μg/mL)[204]
Sulfated Galactofucan LJNF3Brown seaweed Saccharina japonicaLPS-induced zebrafish modelAnti-inflammatory effect (12.5, 25, 50 μg/mL)[205]
Eckmaxol and dieckolBrown seaweed Ecklonia maximaLPS-induced zebrafish modelAnti-inflammatory effect (12.5, 25, 50 μg/mL)[206]
Enzymatic peptide SEPSkipjack Katsuwonus pelamisCuSO4-induced Tg(zlyz: EGFP) zebrafishAnti-inflammatory activity (500 μg/mL)[207]
Micrometam CMicromelum falcatumLPS-induced zebrafish modelAnti-inflammatory activity (10, 50, 200 μM)[208]
Peptides YSQLENEFDR and
YIAEDAER
Marine snailmetronidazole-treated Tg (krt4: NTR-hKikGR)cy17 zebrafishAntioxidant activity (0.77, 7.70, 76.98 and 1.03, 10.36, 103.63 μM)[213]
Frondoplysin AMarine sponge Dysidea frondosametronidazole-treated Tg (krt4: NTR-hKikGR)cy17 zebrafishAntioxidant activity (10, 20, 40 μM)[214]
Peptide AFFPAquacultured flounder fish2,2-azobis-(2-amidinopropane) hydrochloride-induced oxidative damage in a zebrafish modelAntioxidative effects (25, 50, 100 μg/mL)[215]
Seahorse peptide SHPHippocampus
abdominalis
AAPH-induced oxidative stress in the zebrafish modelAntioxidative effects (50, 100, 200 μg/mL)[216]
Phloroglucinol, eckol, dieckol, eckstolonol and triphloroethol ABrown algae Ecklonia cavaAAPH-induced oxidative stress in the zebrafish modelAntioxidative effects (50 μM)[217]
Agaro-oligosaccharides AOAgar of Gracilaria lemaneiformisH2O2-stimulated oxidative stress in zebrafishAntioxidant activity (12.5, 25, 50 μg/mL)[218]
Xyloketal BMangrove
fungus Xylaria sp. (no. 2508)
Phorbol myristate acetate-induced ROS levels in zebrafishAntioxidant activity (20 μM)[219]
(−)-loliolideSargassum horneriAAPH-induced oxidative damage in zebrafish modelsAntioxidant activity (6.25, 12.5, 25 μg/mL)[220]
Protein hydrolysate HPHSeahorses Hippocampus abdominalisAAPH-induced oxidative damage in zebrafishAntioxidant activity (100, 400 μg/mL)[221]
Fucoidan (HFPS-F4)Hizikia fusiformeH2O2-stimulated oxidative stress in zebrafishAntioxidant activity (12.5, 25, 50 μg/mL)[222]

4. Conclusions

Marine natural products hold immense promise in drug discovery, necessitating animal models that simulate human biology and provide high-throughput, accurate screening for identifying bioactive lead molecules. Zebrafish, as a vertebrate model, bridges the gap between cellular and mammalian studies [8,11]. Compared to mammals, zebrafish offer advantages in reproduction, development, and feeding while maintaining high genetic and physiological conservation with humans. Zebrafish disease models allow for an effective exploration of disease mechanisms and solutions, making them valuable for activity screening. With advancements in automation and analysis technologies, zebrafish models enable high-throughput screening and economical evaluation of bioactive compounds with potential human benefits. They are widely used in biomedicine, personal care, functional foods, and natural product activity screening and evaluation [9,10].
This review summarized zebrafish models related to cardiovascular diseases, cancer, metabolic disorders, inflammation, and oxidative stress, emphasizing their use in evaluating marine natural products. Various zebrafish models have been developed, including chemically induced, diet-modified, and fluorescently labeled transgenic strains, which facilitate the observation of bioactive component effects. Among the research areas, our team has primarily focused on cardiovascular diseases, inflammation, and oxidative stress, though zebrafish models are also being increasingly applied in discovering marine bioactive ingredients, showcasing significant potential for future exploration. In addition to the zebrafish models in this review, there are some zebrafish models for neuroprotective activity, safety evaluation, and guidance on the isolation of active natural products [17]. The bioactive constituents obtained from terrestrial sources evaluated by zebrafish models have been applied in clinical research or marketed drugs, such as ferulic acid and danhong extracts. Sodium ferulate and danhong injection are extensively used to treat cardiovascular and cerebrovascular diseases in the clinic, and their proangiogenic activity was evaluated by a zebrafish model [223,224]. However, to our knowledge, there are currently few marine bioactive molecules evaluated by a zebrafish model in clinical research, indicating that zebrafish still have great potential in the exploration of marine bioactive molecules.
The application of the zebrafish model in studying marine bioactive molecules is still relatively limited compared to terrestrial sources, particularly traditional Chinese medicine, and requires more comprehensive and in-depth research. Many zebrafish models have not yet been fully applied to the discovery of marine bioactive molecules, and specific transgenic zebrafish models for certain diseases are still lacking. Therefore, further exploration and optimization of zebrafish models in this field are necessary. The zebrafish model also lacks standardized experimental operations, such as unified environmental conditions, technical procedures, and statistical analysis methods [17]. Establishing standardized experimental criteria and guidelines for each zebrafish model, along with building a database containing multiple models and their corresponding criteria, would greatly assist researchers in selecting appropriate models. Zebrafish models, constructed through gene manipulation techniques like specific gene knockout, overexpression or the introduction of target genes, are essential for studying the mechanisms of marine bioactive molecules and understanding their mechanistic relationships with human biology [8]. Although zebrafish genes are highly similar to those of humans, certain differences in the encoded proteins mean that drugs suitable for zebrafish may not achieve the same effects in humans. Therefore, confirming research results with other model organisms and clinical trials is crucial. Additionally, the larval zebrafish model poses challenges in quantifying drug administration, as substances are typically dissolved in water, making it difficult to measure true absorption and potentially leading to false negatives. More research is needed on bioavailability to overcome these limitations [17]. In conclusion, the potential of zebrafish models in screening marine bioactive molecules is significant, and with further advancements in science and technology, the discovery of marine bioactive molecules will make greater contributions to human medical research.

Author Contributions

Conceptualization, P.L. and Y.Z.; data analysis, D.W. and J.L. (Jibin Liu); writing—original draft preparation, C.L. and J.L. (Jiaxun Li); writing—review and editing, P.L.; funding acquisition, Y.Z. and K.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Key R&D Program of China (2022YFC2804600), the Shandong Provincial Natural Science Foundation (ZR2021ZD29), the Jinan Talent Project for University (202228035), and the Basic and Talent Research Project of the Pilot Project for the Integration of Science, Education and Industry, Qilu University of Technology, Shandong Academy of Sciences (2023RCKY229).

Data Availability Statement

No new data were generated.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Choudhary, A.; Naughton, L.M.; Montánchez, I.; Dobson, A.D.W.; Rai, D.K. Current status and future prospects of marine natural products (MNPs) as antimicrobials. Mar. Drugs 2017, 15, 272. [Google Scholar] [CrossRef] [PubMed]
  2. Lin, Q.F.; Gan, C.F.; Cui, J.G. Advances in the research of bioactive steroids derived from marine microorganisms. Nat. Prod. Res. Dev. 2018, 30, 1266–1273. [Google Scholar]
  3. Barbosa, F.; Pinto, E.; Kijjoa, A.; Pinto, M.; Sousa, E. Targeting antimicrobial drug resistance with marine natural products. Int. J. Antimicrob. Agents 2020, 56, 106005. [Google Scholar] [CrossRef] [PubMed]
  4. Cutolo, E.A.; Campitiello, R.; Caferri, R.; Pagliuca, V.F.; Li, J.; Agathos, S.N.; Cutolo, M. Immunomodulatory compounds from the sea: From the origins to a modern marine pharmacopoeia. Mar. Drugs 2024, 22, 304. [Google Scholar] [CrossRef] [PubMed]
  5. Li, X.; Vitiello, L.; Giannaccare, G. Marine natural products rescuing the eye: A narrative review. Mar. Drugs 2024, 22, 155. [Google Scholar] [CrossRef] [PubMed]
  6. Zhu, W. Efficient discovery and medicinal research of marine natural products. Chin. Tradit. Herb. Drugs 2019, 50, 5645–5652. [Google Scholar]
  7. Ha, Q.; Parveen, S.; Tang, T.; Wei, J.; Huang, Z. Marine natural products in clinical use. Mar. Drugs 2022, 20, 528. [Google Scholar]
  8. Mandrekar, N.; Thakur, N.L. Significance of the zebrafish model in the discovery of bioactive molecules from nature. Biotechnol. Lett. 2009, 31, 171–179. [Google Scholar] [CrossRef] [PubMed]
  9. Zhao, C.; Jia, S.; Zhang, X. Recent advances in pharmaceutical screening using zebrafish as a disease model. Period. Ocean Univ. China 2019, 49, 59–65. [Google Scholar]
  10. West, K.H.; Crawford, A.D. Marine Biodiscovery Goes Deeper: Using In Vivo Bioassays Based on model organisms to identify biomedically relevant marine metabolites. Planta Med. 2016, 82, 754–760. [Google Scholar] [CrossRef]
  11. Parng, C.; Seng, W.L.; Semino, C.; McGrath, P. Zebrafish: A preclinical model for drug screening. Assay Drug Dev. Technol. 2002, 1 Pt 1, 41–48. [Google Scholar] [CrossRef] [PubMed]
  12. Kent, M.L.; Wall, E.S.; Sichel, S.; Watral, V.; Stagaman, K.; Sharpton, T.J.; Guillemin, K. Pseudocapillaria tomentosa, Mycoplasma spp., and intestinal lesions in experimentally infected zebrafish danio rerio. Zebrafish 2021, 18, 207–220. [Google Scholar] [CrossRef] [PubMed]
  13. Hisaoka, K.K. The effects of 2-acetylaminofluorene on the embryonic development of the zebrafish. II. Histochemical studies. Cancer Res. 1958, 18, 664–667. [Google Scholar]
  14. Streisinger, G.; Walker, C.; Dower, N.; Knauber, D.; Singer, F. Production of clones of homozygous diploid zebra fish (Brachydanio rerio). Nature 1981, 291, 293–296. [Google Scholar] [CrossRef] [PubMed]
  15. Streisinger, G.; Singer, F.; Walker, C.; Knauber, D.; Dower, N. Segregation analyses and genecentrome distances in zebrafish. Genetics 1986, 112, 311–319. [Google Scholar] [CrossRef]
  16. Peterson, R.T.; Link, B.A.; Dowling, J.E.; Schreiber, S.L. Small molecule developmental screens reveal the logic and timing of vertebrate development. Proc. Natl. Acad. Sci. USA 2000, 97, 12965–12969. [Google Scholar] [CrossRef] [PubMed]
  17. Lin, F.J.; Li, H.; Wu, D.T.; Zhuang, Q.G.; Li, H.B.; Geng, F.; Gan, R.Y. Recent development in zebrafish model for bioactivity and safety evaluation of natural products. Crit. Rev. Food Sci. Nutr. 2022, 62, 8646–8674. [Google Scholar] [CrossRef] [PubMed]
  18. Kithcart, A.; MacRae, C.A. Using zebrafish for high-throughput screening of novel cardiovascular drugs. JACC Basic Transl. Sci. 2017, 2, 1–12. [Google Scholar] [CrossRef]
  19. Crawford, A.D.; Esguerra, C.V.; de Witte, P.A. Fishing for drugs from nature: Zebrafish as a technology platform for natural product discovery. Planta Med. 2008, 74, 624–632. [Google Scholar] [CrossRef] [PubMed]
  20. Meyers, J.R. Zebrafish: Development of a vertebrate model organism. Curr. Protoc. Essent. Lab. Tech. 2018, 16, 19. [Google Scholar] [CrossRef]
  21. Spence, R.; Gerlach, G.; Lawrence, C.; Smith, C. The behaviour and ecology of the zebrafish, danio rerio. Biol. Rev. Camb. Philos. Soc. 2008, 83, 13–34. [Google Scholar] [CrossRef]
  22. Cueto-Escobedo, J.; German-Ponciano, L.J.; Guillén-Ruiz, G.; Soria-Fregozo, C.; Herrera-Huerta, E.V. Zebrafish as a useful tool in the research of natural products with potential anxiolytic effects. Front. Behav. Neurosci. 2022, 15, 795285. [Google Scholar] [CrossRef]
  23. Zhang, Y.; Xia, Q.; Wang, J.; Zhuang, K.; Jin, H.; Liu, K. Progress in using zebrafish as a toxicological model for traditional chinese medicine. J. Ethnopharmacol. 2022, 282, 114638. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, R.J.; Zhu, J.J.; Zhou, S.H.; Xin, Y.; Xuan, Y.; Li, C. Application of zebrafish model in drug toxicity and safety evaluation. J. Toxicol. 2012, 26, 224–228. [Google Scholar]
  25. Challal, S.; Bohni, N.; Buenafe, O.E.; Esguerra, C.V.; de Witte, P.A.; Wolfender, J.L.; Crawford, A.D. Zebrafish bioassay-guided microfractionation for the rapid in vivo identification of pharmacologically active natural products. Chimia 2012, 66, 229–232. [Google Scholar] [CrossRef]
  26. Lin, S.; Zhao, Y.; Nel, A.E.; Lin, S. Zebrafish: An in vivo model for nano EHS studies. Small 2013, 9, 1608–1618. [Google Scholar] [CrossRef] [PubMed]
  27. Bohni, N.; Cordero-Maldonado, M.L.; Maes, J.; Siverio-Mota, D.; Marcourt, L.; Munck, S.; Kamuhabwa, A.R.; Moshi, M.J.; Esguerra, C.V.; de Witte, P.A.; et al. Integration of microfractionation, qNMR and zebrafish screening for the in vivo bioassay-guided isolation and quantitative bioactivity analysis of natural products. PLoS ONE 2013, 8, e64006. [Google Scholar] [CrossRef] [PubMed]
  28. Howe, K.; Clark, M.D.; Torroja, C.F.; Torrance, J.; Berthelot, C.; Muffato, M.; Collins, J.E.; Humphray, S.; McLaren, K.; Matthews, L.; et al. The zebrafish reference genome sequence and its relationship to the human genome. Nature 2013, 7446, 496–505. [Google Scholar] [CrossRef]
  29. Giacomotto, J.; Ségalat, L. High-throughput screening and small animal models, where are we? Br. J. Pharmacol. 2010, 160, 204–216. [Google Scholar] [CrossRef]
  30. Liu, J.T.; Tong, K.; Cui, J.R. Application of zebrafish in tumor pharmacology. Chin. Pharm. J. 2010, 5, 1800–1804. [Google Scholar]
  31. Maximino, C.; da Silva, A.W.; Araújo, J.; Lima, M.G.; Miranda, V.; Puty, B.; Benzecry, R.; Picanço-Diniz, D.L.; Gouveia, A.; Oliveira, K.R., Jr.; et al. Fingerprinting of psychoactive drugs in zebrafish anxiety-like behaviors. PLoS ONE 2014, 9, e103943. [Google Scholar] [CrossRef] [PubMed]
  32. Li, Z.; Zheng, W.; Wang, Z.; Zeng, Z.; Zhan, H.; Li, C.; Zhou, L.; Yan, C.; Spitsbergen, J.M.; Gong, Z. A transgenic zebrafish liver tumor model with inducible Myc expression reveals conserved Myc signatures with mammalian liver tumors. Dis. Models Mech. 2013, 6, 414–423. [Google Scholar] [CrossRef] [PubMed]
  33. Kundap, U.P.; Kumari, Y.; Othman, I.; Shaikh, M.F. Zebrafish as a model for epilepsy-induced cognitive dysfunction: A pharmacological, biochemical and behavioral approach. Front. Pharmacol. 2017, 8, 515–851. [Google Scholar] [CrossRef] [PubMed]
  34. Hammond, C.L.; Moro, E. Using transgenic reporters to visualize bone and cartilage signaling during development in vivo. Front. Endocrinol. 2012, 3, 91. [Google Scholar] [CrossRef]
  35. Peng, W.; Zhang, W.J.; Xue, Y. Research progress of zebrafish as a model for skeletal diseases. Acta Lab. Anim. Sci. Sin. 2019, 27, 248–253. [Google Scholar]
  36. Xin, S.C.; Zhao, Y.Q.; Li, S.; Lin, S.; Zhong, H.B. Application of zebrafish models in drug screening. Hereditas 2012, 34, 1144–1152. [Google Scholar] [PubMed]
  37. Bournele, D.; Beis, D. Zebrafish models of cardiovascular disease. Heart Fail. Rev. 2016, 21, 803–813. [Google Scholar] [CrossRef]
  38. Zhang, Y.P. Establishment of Thrombocyte Specific Transgenic Line and Expression Analysis and Generation of Gene Knockout Mutant with CRISPR/Cas9 System in Zebrafish. Master’s Thesis, Southern Medical University, Guangzhou, China, 2016. [Google Scholar]
  39. Qu, Y.G. Establishment and Preliminary Functional Study of Several Zebrafish Transgenic Lines. Master’s Thesis, Hunan Normal University, Changsha, China, 2014. [Google Scholar]
  40. Zhang, S.H.; Zhao, C.T.; Fen, X.Z. Research progress of zebrafish cancer model in tumor precision medicine. J. Clin. Pathol. Res. 2021, 41, 2193–2200. [Google Scholar]
  41. Lv, P.; Ma, D.; Gao, S.; Zhang, Y.; Bae, Y.K.; Liang, G.; Gao, S.; Choi, J.H.; Kim, C.H.; Wang, L.; et al. Generation of foxn1/casper mutant zebrafish for allograft and xenograft of normal and malignant cells. Stem Cell Rep. 2020, 15, 749–760. [Google Scholar] [CrossRef]
  42. Kettborough, R.N.; Busch-Nentwich, E.M.; Harvey, S.A.; Dooley, C.M.; de Bruijn, E.; van Eeden, F.; Sealy, I.; White, R.J.; Herd, C.; Nijman, I.J.; et al. A systematic genome-wide analysis of zebrafish protein-coding gene function. Nature 2023, 496, 494–497. [Google Scholar] [CrossRef]
  43. Gut, P.; Reischauer, S.; Stainier, D.Y.R.; Arnaout, R. Little Fish, Big Data: Zebrafish as a Model for Cardiovascular and Metabolic Disease. Physiol. Rev. 2017, 97, 889–938. [Google Scholar] [CrossRef] [PubMed]
  44. Kithcart, A.P.; MacRae, C.A. Zebrafish assay development for cardiovascular disease mechanism and drug discovery. Prog. Biophys. Mol. Biol. 2018, 138, 126–131. [Google Scholar] [CrossRef] [PubMed]
  45. Bakkers, J. Zebrafish as a model to study cardiac development and human cardiac disease. Cardiovasc. Res. 2011, 91, 279–288. [Google Scholar] [CrossRef] [PubMed]
  46. Farrell, A.P.; Pieperhoff, S. Design and physiology of the heart I cardiac anatomy in fishes. In Encyclopedia of Fish Physiology; Academic Press: Cambridge, MA, USA, 2011; pp. 998–1005. [Google Scholar]
  47. Zakaria, Z.Z.; Benslimane, F.M.; Nasrallah, G.K.; Shurbaji, S.; Younes, N.N.; Mraiche, F.; Da’as, S.I.; Yalcin, H.C. Using zebrafish for investigating the molecular mechanisms of drug-induced cardiotoxici. BioMed Res. Int. 2018, 2018, 1642684. [Google Scholar] [CrossRef] [PubMed]
  48. Ran, K.K.; Zhen, R.F.; XIA, Q.; Zhang, C.Q.; Ma, R.J.; Liu, K.Y.; Zhang, Y.; Xing, J.G. Application of model organism-zebrafish in cardiac function evaluation. Drug Eval. Res. 2021, 44, 1581–1587. [Google Scholar]
  49. Li, P.H.; Zhang, M.Q.; Xie, D.X.; Zhang, X.M.; Zhang, S.S.; Gao, F.Y.; Wang, Y.L.; Hsiao, C.D.; Li, X.B.; Liu, K.C. Characterization and bioactivities of phospholipids from squid viscera and gonads using ultra-performance liquid chromatography-Q-exactive orbitrap/mass spectrometry-based lipidomics and zebrafish models. Food Funct. 2021, 12, 7986–7996. [Google Scholar] [CrossRef] [PubMed]
  50. Zhu, Y.Q.; Li, P.H.; Meng, R.H.; Li, X.B.; Qiu, Y.Z.; Wang, L.Z.; Zhang, S.S.; Zhang, X.M.; Lin, H.W.; Zhai, H.B.; et al. Lipid profiles of the heads of four shrimp species by UPLC-Q-Exactive Orbitrap/MS and their cardiovascular activities. Molecules 2022, 27, 350. [Google Scholar] [CrossRef] [PubMed]
  51. Xie, D.X.; Li, P.H.; Zhu, Y.Q.; Zhang, M.Q.; He, J.W.; Liu, K.C.; Lin, H.W.; Zhai, H.B.; Li, X.B.; Ma, Y.H. Comparative bioactivity profile of phospholipids from three marine byproducts based on the zebrafish model. J. Food Biochem. 2022, 46, e14229. [Google Scholar] [CrossRef]
  52. Zhang, M.Q.; Li, P.H.; Wang, F.X.; Zhang, S.S.; Li, H.N.; Zhang, Y.; Wang, X.M.; Liu, K.C.; Li, X.B. Separation, identification and cardiovascular activities of phospholipid classes from the head of Penaeus vannamei by lipidomics and zebrafish models. Food Funct. 2021, 12, 2282–2291. [Google Scholar] [CrossRef] [PubMed]
  53. Liao, Q.; Gong, G.; Siu, S.W.I.; Wong, C.T.T.; Yu, H.; Tse, Y.C.; Rádis-Baptista, G.; Lee, S.M. A novel shk-like toxic peptide from the transcriptome of the cnidarian Palythoa caribaeorum displays neuroprotection and cardioprotection in zebrafish. Toxins 2018, 10, 238. [Google Scholar] [CrossRef] [PubMed]
  54. Pott, A.; Shahid, M.; Köhler, D.; Pylatiuk, C.; Weinmann, K.; Just, S.; Rottbauer, W. Therapeutic chemical screen identifies phosphatase inhibitors to reconstitute PKB phosphorylation and cardiac contractility in ILK-Deficient zebrafish. Biomolecules 2018, 8, 153. [Google Scholar] [CrossRef]
  55. Hu, X.Y.; You, H.M.; Ren, C.Z.; Zhang, Y.D.; Liang, C. Application of zebrafish in drug screening research for cardiovascular diseases. Int. J. Cardiovasc. Dis. 2021, 48, 156–159. [Google Scholar]
  56. Chen, Y.; Xu, H.; Wang, Y.J.; Shi, Q.; Liang, Q.Q. Application of zebrafish model in study of traditional chinese medicine regulating angiogenesis. Chin. Arch. Tradit. Chin. Med. 2023, 41, 46–50. [Google Scholar]
  57. Wilkinson, R.N.; van Eeden, F.J. The zebrafish as a model of vascular development and disease. Prog. Mol. Biol. Transl. Sci. 2014, 124, 93–122. [Google Scholar]
  58. Matsuoka, R.L.; Stainier, D.Y.R. Recent insights into vascular development from studies in zebrafish. Curr. Opin. Hematol. 2018, 25, 204–211. [Google Scholar] [CrossRef] [PubMed]
  59. Lu, X.L.; Xu, Z.L.; Yao, X.L.; Su, F.J.; Ye, C.H.; Li, J.; Lin, Y.C.; Wang, G.L.; Zeng, J.S.; Huang, R.X.; et al. Marine cyclotripeptide X-13 promotes angiogenesis in zebrafish and human endothelial cells via PI3K/Akt/eNOS signaling pathways. Mar. Drugs 2012, 10, 1307–1320. [Google Scholar] [CrossRef]
  60. Li, P.; Zhang, M.; Li, H.; Wang, R.; Hou, H.; Li, X.; Liu, K.; Chen, H. New prenylated indole homodimeric and pteridine alkaloids from the marine-derived fungus Aspergillus austroafricanus Y32-2. Mar. Drugs 2021, 19, 98. [Google Scholar] [CrossRef] [PubMed]
  61. Fan, Y.Q.; Li, P.H.; Chao, Y.X.; Chen, H.; Du, N.; He, Q.X.; Liu, K.C. Alkaloids with cardiovascular effects from the marine-derived fungus Penicillium expansum Y32. Mar. Drugs 2015, 13, 6489–6504. [Google Scholar] [CrossRef]
  62. 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 J. Biosci. 2023, 78, 345–352. [Google Scholar] [CrossRef] [PubMed]
  63. Fan, Y.; Jiang, C.; Zhang, Y.; Ma, Z.; Li, P.; Guo, L.; Feng, T.; Zhou, L.; Xu, L. Pro-angiogenic new chloro-azaphilone derivatives from the hadal trench-derived fungus Chaetomium globosum YP-106. Front. Microbiol. 2022, 13, 943452. [Google Scholar] [CrossRef]
  64. Yan, L.H.; Li, P.H.; Li, X.M.; Yang, S.Q.; Liu, K.C.; Zhang, Y.; Wang, B.G.; Li, X. Bialorastins A–F, highly oxygenated and polycyclic andrastin-type meroterpenoids with proangiogenic activity from the deep-sea cold-seep-derived fungus Penicillium bialowiezense CS-283. Bioorganic Chem. 2024, 143, 107073. [Google Scholar] [CrossRef] [PubMed]
  65. Han, X.; Tang, X.; Luo, X.; Sun, C.; Liu, K.; Zhang, Y.; Li, P.; Li, G. Isolation and identification of three new sterigmatocystin derivatives from the fungus Aspergillus versicolor guided by molecular networking approach. Chem. Biodivers. 2020, 17, e2000208. [Google Scholar] [CrossRef]
  66. Li, T.; Tang, X.; Luo, X.; Wang, Q.; Liu, K.; Zhang, Y.; de Voogd, N.J.; Yang, J.; Li, P.; Li, G. Agelanemoechine, a dimeric bromopyrrole alkaloid with a Pro-Angiogenic effect from the south china sea sponge Agelas nemoechinata. Org. Lett. 2019, 21, 9483–9486. [Google Scholar] [CrossRef] [PubMed]
  67. Liu, M.; Li, P.; Tang, X.; Luo, X.; Liu, K.; Zhang, Y.; Wang, Q.; Li, G. Lemnardosinanes A–I: New bioactive sesquiterpenoids from soft coral Lemnalia sp. J. Org. Chem. 2021, 86, 970–979. [Google Scholar] [CrossRef]
  68. Miao, X.; Hong, L.; Ju, Z.; Liu, H.; Shang, R.; Li, P.; Liu, K.; Cheng, B.; Jiao, W.; Xu, S.; et al. Marchaetoglobins A-D: Four cytochalasans with proangiogenic activity from the marine-sponge-associated fungus Chaetomium globosum 162105. ACS Omega 2024, 9, 22450–22458. [Google Scholar] [CrossRef] [PubMed]
  69. Yan, L.H.; Li, P.H.; Li, X.M.; Yang, S.Q.; Liu, K.C.; Wang, B.G.; Li, X. Chevalinulins A and B, proangiogenic alkaloids with a spiro[bicyclo[2.2.2]octane-diketopiperazine] skeleton from deep-sea cold-seep-derived fungus Aspergillus chevalieri CS-122. Org. Lett. 2022, 24, 2684–2688. [Google Scholar] [CrossRef]
  70. Wang, Q.; Tang, X.; Liu, H.; Luo, X.; Sung, P.J.; Li, P.L.; Li, G.Q. Clavukoellians G–K, new nardosinane and aristolane sesquiterpenoids with angiogenesis promoting activity from the marine soft coral Lemnalia sp. Mar. Drugs 2020, 18, 171. [Google Scholar] [CrossRef] [PubMed]
  71. Fan, Y.Q.; Jiang, C.; Zhang, Y.J.; Ma, Z.H.; Li, P.H.; Guo, L.Z.; Feng, T.; Zhou, L.M.; Xu, L.L. A new chloro-azaphilone derivative with pro-angiogenesis activity from the hadal trench-derived fungus Chaetomium globosum YP-106. J. Oceanol. Limnol. 2023, 41, 1145–1151. [Google Scholar] [CrossRef]
  72. Li, Z.; Wu, N.; Wang, J.; Yue, Y.; Geng, L.; Zhang, Q. Low molecular weight fucoidan alleviates cerebrovascular damage by promoting angiogenesis in type 2 diabetes mice. Int. J. Biol. Macromol. 2022, 217, 345–355. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, Q.; Xu, N.; Zhao, C.; He, Y.L.; Kam, S.H.T.; Wu, X.; Huang, P.; Yang, M.; Wong, C.T.T.; Radis-Baptista, G.; et al. A new invertebrate NPY-like polypeptide, ZoaNPY, from the zoanthus sociatus, as a novel ligand of human NPY Y2 receptor rescues vascular insufficiency via PLC/PKC and Src- FAK-dependent signaling pathways. Pharmacol. Res. 2024, 203, 107173. [Google Scholar] [CrossRef]
  74. Feng, T.; Wu, R.; Wang, Y.; Wang, P.; Zhou, L.; Wang, C.; Kong, F. Proangiogenic azaphilones from the marine-derived fungus neopestalotiopsis sp. HN-1-6. Mar. Drugs 2024, 22, 241. [Google Scholar] [CrossRef]
  75. Tulotta, C.; He, S.; van der Ent, W.; Chen, L.; Groenewoud, A.; Spaink, H.P.; Snaar-Jagalska, B.E. Imaging cancer angiogenesis and metastasis in a zebrafish embryo model. Adv. Exp. Med. Biol. 2016, 916, 239–263. [Google Scholar] [PubMed]
  76. García-Caballero, M.; Cañedo, L.; Fernández-Medarde, A.; Medina, M.Á.; Quesada, A.R. The marine fungal metabolite, AD0157, inhibits angiogenesis by targeting the akt signaling pathway. Mar. Drugs 2014, 12, 279–299. [Google Scholar] [CrossRef]
  77. Liu, J.X.; Luo, M.Q.; Xia, M.; Wu, Q.; Long, S.M.; Hu, Y.; Gao, G.C.; Yao, X.L.; He, M.; Su, H.; et al. Marine compound catunaregin inhibits angiogenesis through the modulation of phosphorylation of akt and eNOS in vivo and in vitro. Mar. Drugs 2014, 12, 2790–2801. [Google Scholar] [CrossRef] [PubMed]
  78. Qi, X.; Liu, G.; Qiu, L.; Lin, X.; Liu, M. Marine bromophenol bis(2,3-dibromo-4,5-dihydroxybenzyl) ether, represses angiogenesis in HUVEC cells and in zebrafish embryos via inhibiting the VEGF signal systems. Biomed. Pharmacother. 2015, 75, 58–66. [Google Scholar] [CrossRef] [PubMed]
  79. Cheng, S.Y.; Chen, N.F.; Lin, P.Y.; Su, J.H.; Chen, B.H.; Kuo, H.M.; Sung, C.S.; Sung, P.J.; Wen, Z.H.; Chen, W.F. Anti-invasion and antiangiogenic effects of stellettin B through Inhibition of the Akt/Girdin signaling pathway and VEGF in glioblastoma cells. Cancers 2019, 11, 220. [Google Scholar] [CrossRef] [PubMed]
  80. Fernando, K.H.N.; Yang, H.W.; Jiang, Y.; Jeon, Y.J.; Ryu, B. Ishige okamurae extract and its constituent ishophloroglucin a attenuated in vitro and in vivo high glucose-induced angiogenesis. Int. J. Mol. Sci. 2019, 20, 5542. [Google Scholar] [CrossRef]
  81. Liu, G.; Liu, M.; Wei, J.; Huang, H.; Zhang, Y.; Zhao, J.; Xiao, L.; Wu, N.; Zheng, L.; Lin, X. CS5931, a novel polypeptide in ciona savignyi, represses angiogenesis via inhibiting vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs). Mar. Drugs 2014, 12, 1530–1544. [Google Scholar] [CrossRef]
  82. Fernando, K.H.N.; Yang, H.W.; Jiang, Y.; Jeon, Y.J.; Ryu, B. Diphlorethohydroxycarmalol isolated from ishige okamurae represses high glucose-induced angiogenesis in vitro and in vivo. Mar. Drugs 2018, 16, 375. [Google Scholar] [CrossRef] [PubMed]
  83. Guo, Z.; Shi, H.; Li, C.; Luo, Y.; Bi, S.; Yu, R.; Wang, H.; Liu, W.; Zhu, J.; Huang, W.; et al. Identification and characterization of a novel protein ASP-3 purified from arca subcrenata and its antitumor mechanism. Mar. Drugs 2019, 17, 528. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, G.; Kuang, S.; Wu, S.; Jin, W.; Sun, C. A novel polysaccharide from Sargassum integerrimum induces apoptosis in A549 cells and prevents angiogensis in vitro and in vivo. Sci. Rep. 2016, 6, 26722. [Google Scholar] [CrossRef]
  85. Guo, X.; Fan, A.; Qi, X.; Liu, D.; Huang, J.; Lin, W. Indoloquinazoline alkaloids suppress angiogenesis and inhibit metastasis of melanoma cells. Bioorganic Chem. 2023, 141, 106873. [Google Scholar] [CrossRef] [PubMed]
  86. Hwang, J.; Yang, H.W.; Lu, Y.A.; Je, J.G.; Lee, H.G.; Fernando, K.H.N.; Jeon, Y.J.; Ryu, B. Phloroglucinol and dieckol isolated from ecklonia cava suppress impaired diabetic angiogenesis; a study of in-vitro and in-vivo. Biomed. Pharmacother. 2021, 138, 111431. [Google Scholar] [CrossRef]
  87. Hsu, S.Y.; Wen, Z.H.; Shih, P.C.; Kuo, H.M.; Lin, S.C.; Liu, H.T.; Lee, Y.H.; Wang, Y.J.; Chen, W.F.; Chen, N.F. Sinularin induces oxidative stress-mediated apoptosis and mitochondrial dysfunction, and inhibits angiogenesis in glioblastoma cells. Antioxidants 2022, 11, 1433. [Google Scholar] [CrossRef] [PubMed]
  88. Song, Y.C.; Wu, B.J.; Chiu, C.C.; Chen, C.L.; Zhou, J.Q.; Liang, S.R.; Duh, C.Y.; Sung, P.J.; Wen, Z.H.; Wu, C.Y. Coral-derived natural marine compound GB9 impairs vascular development in zebrafish. Int. J. Mol. Sci. 2017, 18, 1696. [Google Scholar] [CrossRef] [PubMed]
  89. García-Caballero, M.; Marí-Beffa, M.; Cañedo, L.; Medina, M.Á.; Quesada, A.R. Toluquinol, a marine fungus metabolite, is a new angiosuppresor that interferes with the akt pathway. Biochem. Pharmacol. 2013, 85, 1727–1740. [Google Scholar] [CrossRef]
  90. Carrillo, P.; Martínez-Poveda, B.; Cheng-Sánchez, I.; Guerra, J.; Tobia, C.; López-Romero, J.M.; Sarabia, F.; Medina, M.Á.; Quesada, A.R. Exploring the antiangiogenic potential of solomonamide a bioactive precursors: In vitro and in vivo evidences of the inhibitory activity of Solo F-OH during angiogenesis. Mar. Drugs 2019, 17, 228. [Google Scholar] [CrossRef] [PubMed]
  91. Wrasidlo, W.; Mielgo, A.; Torres, V.A.; Barbero, S.; Stoletov, K.; Suyama, T.L.; Klemke, R.L.; Gerwick, W.H.; Carson, D.A.; Stupack, D.G. The marine lipopeptide somocystinamide A triggers apoptosis via caspase 8. Proc. Natl. Acad. Sci. USA 2008, 105, 2313–2318. [Google Scholar] [CrossRef] [PubMed]
  92. Bae, H.; Lee, J.Y.; Yang, C.; Song, G.; Lim, W. Fucoidan derived from Fucus vesiculosus inhibits the development of human ovarian cancer via the disturbance of calcium homeostasis, endoplasmic reticulum stress, and angiogenesis. Mar. Drugs 2020, 18, 45. [Google Scholar] [CrossRef] [PubMed]
  93. Lin, S.W.; Huang, S.C.; Kuo, H.M.; Chen, C.H.; Ma, Y.L.; Chu, T.H.; Bee, Y.S.; Wang, E.M.; Wu, C.Y.; Sung, P.J.; et al. Coral-derived compound WA-25 inhibits angiogenesis by attenuating the VEGF/VEGFR2 signaling pathway. Mar. Drugs 2015, 13, 861–878. [Google Scholar] [CrossRef]
  94. Dong, S.T.; Chen, Z.Y.; Wang, L.; Liu, Y.K.; Stagos, D.; Lin, X.K.; Liu, M. Marine bromophenol bis(2,3,6-Tribromo-4,5-Dihydroxybenzyl)ether inhibits angiogenesis in human umbilical vein endothelial cells and reduces vasculogenic mimicry in human lung cancer A549 cells. Mar. Drugs 2021, 19, 641. [Google Scholar] [CrossRef]
  95. Zeng, Q.; Chen, Y.C.; Wang, J.F.; Shi, X.F.; Che, Y.H.; Chen, X.Y.; Zhong, W.M.; Zhang, W.M.; Wei, X.Y.; Wang, F.Z.; et al. Diverse secondary metabolites from the coral-derived fungus Aspergillus hiratsukae SCSIO 5Bn1003. Mar. Drugs 2022, 20, 150. [Google Scholar] [CrossRef] [PubMed]
  96. Song, N.; Tang, Y.; Wang, Y.; Guan, X.; Yu, W.; Jiang, T.; Lu, L.; Gu, Y. A sirt6 inhibitor, marine-derived pyrrole-pyridinimidazole derivative 8a, suppresses angiogenesis. Mar. Drugs 2023, 21, 517. [Google Scholar] [CrossRef] [PubMed]
  97. 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]
  98. Nagabhishek, S.N.; Madan Kumar, A.; B, S.; Balakrishnan, A.; Katakia, Y.T.; Chatterjee, S.; Nagasundaram, N. A marine sponge associated fungal metabolite monacolin X suppresses angiogenesis by down regulating VEGFR2 signaling. RSC Adv. 2019, 9, 26646–26667. [Google Scholar] [CrossRef] [PubMed]
  99. Ratnayake, R.; Gunasekera, S.P.; Ma, J.J.; Dang, L.H.; Carney, T.J.; Paul, V.J.; Luesch, H. Dolastatin 15 from a marine cyanobacterium suppresses HIF-1α mediated cancer cell viability and vascularization. ChemBioChem 2020, 21, 2356–2366. [Google Scholar] [CrossRef]
  100. Long, W.; Wang, M.; Luo, X.; Huang, G.; Chen, J. Murrangatin suppresses angiogenesis induced by tumor cell-derived media and inhibits AKT activation in zebrafish and endothelial cells. Drug Des. Dev. Ther. 2018, 12, 3107–3115. [Google Scholar] [CrossRef] [PubMed]
  101. Lang, M.R.; Gihr, G.; Gawaz, M.P.; Müller, I.I. Hemostasis in Danio rerio: Is the zebrafish a useful model for platelet research? J. Thromb. Haemost. 2010, 8, 1159–1169. [Google Scholar] [CrossRef]
  102. Thijs, T.; Deckmyn, H.; Broos, K. Model systems of genetically modified platelets. Blood 2012, 119, 1634–1642. [Google Scholar] [CrossRef]
  103. Weyand, A.C.; Shavit, J.A. Zebrafish as a model system for the study of hemostasis and thrombosis. Curr. Opin. Hematol. 2014, 21, 418–422. [Google Scholar] [CrossRef] [PubMed]
  104. Zhu, X.Y.; Liu, H.C.; Guo, S.Y.; Xia, B.; Song, R.S.; Lao, Q.C.; Xuan, Y.X.; Li, C.Q. A zebrafish thrombosis model for assessing antithrombotic drugs. Zebrafish 2016, 13, 335–344. [Google Scholar] [CrossRef]
  105. Rajagopal, R.E.; Balasubramanian, M.; Kalyanaraman, S. Phenylhydrazine hydrochloride induced dosedependent embryo cytotoxicity in zebrafish. Bioinformation 2019, 15, 255–260. [Google Scholar] [CrossRef]
  106. Gao, P.Y.; Li, L.Z.; Liu, K.C.; Sun, C.; Sun, X.; Wu, Y.N.; Song, S.J. Natural terpenoid glycosides with in vitro/vivo antithrombotic profiles from the leaves of crataegus pinnatifida. RSC Adv. 2019, 7, 48466–48474. [Google Scholar] [CrossRef]
  107. Mo, C.L.; Li, J.; Wang, J.Z.; Liu, X.; Lin, S.H. Applicability of a zebrafish thrombosis model in screening active ingredients of traditional Chinese medicine. Shandong Sci. 2021, 34, 52–59. [Google Scholar]
  108. Shi, Y.P.; Zhang, Y.G.; Li, H.N.; Kong, H.T.; Zhang, S.S.; Zhang, X.M.; Li, X.B.; Liu, K.C.; Han, L.W.; Tian, Q.P. Discovery and identification of antithrombotic chemical markers in gardenia fructus by herbal metabolomics and zebrafish model. J. Ethnopharmacol. 2020, 253, 112679. [Google Scholar] [CrossRef]
  109. Li, J.; Liu, H.; Yang, Z.; Yu, Q.; Zhao, L.; Wang, Y. Synergistic effects of cryptotanshinone and senkyunolide I in guanxinning tablet against endogenous thrombus formation in zebrafish. Front. Pharmacol. 2021, 11, 622787. [Google Scholar] [CrossRef]
  110. Xin, S.S.; Zhang, M.Q.; Li, P.H.; Wang, L.Z.; Zhang, X.M.; Zhang, S.S.; Mu, Z.Q.; Lin, H.W.; Li, X.B.; Liu, K.C. Marine-fungus-derived natural compound 4-Hydroxyphenylacetic acid induces autophagy to exert antithrombotic effects in zebrafish. Mar. Drugs 2024, 22, 148. [Google Scholar] [CrossRef] [PubMed]
  111. Wang, C.L.; Zhang, J.R.; Jin, T.Y.; Shi, X.; Wang, M.F.; Zhang, W.J.; Zong, Y.; Li, G.Q.; Li, P.L. Sarcoeleganolides H-K, new anti-thrombotic cembranes from soft coral Sarcophyton elegans. Nat. Prod. Res. 2024, 38, 3597–3604. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, C.; Zhang, J.; Gan, Y.; Wang, M.F.; Li, X.L.; Liu, X.H.; Shi, X.; Mi, Y.; Liu, K.C.; Zhang, Y.; et al. Sarcoelegans A-H, eight undescribed cembranes with anti-inflammatory and anti-thrombotic activities from the south china sea soft coral sarcophyton elegans. Phytochemistry 2023, 207, 113578. [Google Scholar] [CrossRef]
  113. Mi, Y.; Han, X.; Yu, X.; Li, L.; Tang, X.; Li, G. Sarcocinerenolides A, an open-loop decarbonizing cembranolide, and sarcocinerenolides B-I, eight polyoxygenated cembranolides with anti-thrombotic activity from the south china sea soft coral sarcophyton cinereum. Phytochemistry 2024, 223, 114109. [Google Scholar] [CrossRef] [PubMed]
  114. Yang, Y.; Qin, L.; Lu, X.; Lu, L.; Mao, W. A pyruvylated and sulfated galactan from the green alga dictyosphaeria cavernosa: Structure, anticoagulant property and inhibitory effect on zebrafish thrombosis. Carbohydr. Polym. 2024, 324, 121492. [Google Scholar] [CrossRef]
  115. Li, L.; Qiu, R.L.; Wei, K.F.; Sun, D.D. Advances in zebrafish models of tumor. Chin. Pharmacol. Bull. 2022, 38, 168–172. [Google Scholar]
  116. Wu, Z.B.; Zhang, H.H.; Li, R.X.; Sun, N.Y.; Liu, C.X. Progress in the application of zebrafish model in cancer research. China Med. Her. Vol. 2023, 20, 45–48. [Google Scholar]
  117. Raby, L.; Völkel, P.; Le Bourhis, X.; Angrand, P.O. Genetic engineering of zebrafish in cancer research. Cancers 2020, 12, 2168. [Google Scholar] [CrossRef] [PubMed]
  118. Le, X.N.; Zon, L.I. Potential of zebrafish for anticancer drug screening. Expert Opin. Drug Discov. 2008, 3, 1451–1460. [Google Scholar]
  119. Callahan, S.J.; Tepan, S.; Zhang, Y.M.; Lindsay, H.; Burger, A.; Campbell, N.R.; Kim, I.S.; Hollmann, T.J.; Studer, L.; Mosimann, C.; et al. Cancer modeling by transgene electroporation in adult zebrafish (TEAZ). Dis. Model Mech. 2018, 11, 034561. [Google Scholar] [CrossRef] [PubMed]
  120. Wu, S.Y.; Yang, W.Y.; Cheng, C.C.; Hsiao, M.C.; Tsai, S.L.; Lin, H.K.; Lin, K.H.; Yuh, C.H. Low molecular weight fucoidan prevents radiation-induced fibrosis and secondary tumors in a zebrafish model. Cancers 2020, 12, 1608. [Google Scholar] [CrossRef] [PubMed]
  121. Cao, X.Z.; Zhang, B.Q.; Wang, C.F.; Yin, J.N.; Haider, W.; Said, G.; Wei, M.Y.; Lu, L. A terphenyllin derivative CHNQD-00824 from the marine compound library induced DNA damage as a potential anticancer agent. Mar. Drugs 2023, 21, 512. [Google Scholar] [CrossRef] [PubMed]
  122. Shen, Y.; Sheng, R.; Guo, R. Application of zebrafish as a model for anti-cancer activity evaluation and toxicity testing of natural products. Pharmaceuticals 2023, 16, 827. [Google Scholar] [CrossRef] [PubMed]
  123. Fazio, M.; Ablain, J.; Chuan, Y.; Langenau, D.M.; Zon, L.I. Zebrafish patient avatars in cancer biology and precision cancer therapy. Nat. Rev. Cancer 2020, 20, 263–273. [Google Scholar] [CrossRef]
  124. Letrado, P.; de Miguel, I.; Lamberto, I.; Díez-Martínez, R.; Oyarzabal, J. Zebrafish: Speeding up the cancer drug discovery process. Cancer Res. 2018, 78, 6048–6058. [Google Scholar] [CrossRef]
  125. Osmani, N.; Goetz, J.G. Multiscale imaging of metastasis in zebrafish. Trends Cancer 2019, 5, 766–778. [Google Scholar] [CrossRef] [PubMed]
  126. Gutiérrez-Lovera, C.; Vázquez-Ríos, A.J.; Guerra-Varela, J.; Sánchez, L.; de la Fuente, M. The potential of zebrafish as a model organism for improving the translation of genetic anticancer nanomedicines. Genes 2017, 8, 349. [Google Scholar] [CrossRef] [PubMed]
  127. Xu, S.; Jiang, W.Q.; Kuang, Y.M.; Xu, W.F.; Jin, P.F. Research advances on zebrafish models in the application of drug screening for anti-tumorconstituents. Northwest Pharm. J. 2018, 33, 422–425. [Google Scholar]
  128. Hung, S.Y.; Chen, W.F.; Lee, Y.C.; Su, J.H.; Juan, Y.S.; Lin, I.P.; Zhang, Y.H.; Chang, M.K.; Lin, M.Y.; Chen, C.Y.; et al. Rhopaloic acid a induces apoptosis, autophagy and MAPK activation through ROS-mediated signaling in bladder cancer. Phytomedicine 2021, 92, 153720. [Google Scholar] [CrossRef]
  129. Chen, W.F.; Tsai, S.C.; Zhang, Y.H.; Chang, H.M.; Wu, W.J.; Su, J.H.; Wu, B.N.; Chen, C.Y.; Lin, M.Y.; Chen, H.L.; et al. Rhopaloic acid a triggers mitochondria damage-induced apoptosis in oral cancer by JNK/BNIP3/Nix-mediated mitophagy. Phytomedicine 2024, 132, 155855. [Google Scholar] [CrossRef]
  130. Chang, C.F.; Chang, P.C.; Lee, Y.C.; Pan, C.Y.; Chang, H.M.; Wu, W.J.; Lin, M.Y.; Chen, C.Y.; Wen, Z.H.; Lee, C.H. The antimicrobial peptide tilapia piscidin 4 induced the apoptosis of bladder cancer through ERK/SIRT1/PGC-1alpha signaling pathway. Probiotics Antimicrob. Proteins, 2024; online ahead of print. [Google Scholar]
  131. Roel, M.; Rubiolo, J.A.; Guerra-Varela, J.; Silva, S.B.; Thomas, O.P.; Cabezas-Sainz, P.; Sánchez, L.; López, R.; Botana, L.M. Marine guanidine alkaloids crambescidins inhibit tumor growth and activate intrinsic apoptotic signaling inducing tumor regression in a colorectal carcinoma zebrafish xenograft model. Oncotarget 2016, 7, 83071–83087. [Google Scholar] [CrossRef] [PubMed]
  132. Yue, Z.Q.; Wang, A.Y.; Zhu, Z.J.; Tao, L.; Li, Y.; Zhou, L.; Chen, W.X.; Lu, Y. Holothurian glycosaminoglycan inhibits metastasis via inhibition of P-selectin in B16F10 melanoma cells. Mol. Cell. Biochem. 2015, 410, 143–154. [Google Scholar] [CrossRef] [PubMed]
  133. Kim, E.A.; Lee, J.H.; Heo, S.J.; Jeon, Y.J. Saringosterol acetate isolated from Hizikia fusiforme, an edible brown alga, suppressed hepatocellular carcinoma growth and metastasis in a zebrafish xenograft model. Chem. Biol. Interact. 2021, 335, 109362. [Google Scholar] [CrossRef] [PubMed]
  134. Jinendiran, S.; Teng, W.; Dahms, H.U.; Liu, W.; Ponnusamy, V.K.; Chiu, C.C.; Kumar, B.S.D.; Sivakumar, N. Induction of mitochondria-mediated apoptosis and suppression of tumor growth in zebrafish xenograft model by cyclic dipeptides identified from Exiguobacterium acetylicum. Sci. Rep. 2020, 10, 13721. [Google Scholar] [CrossRef] [PubMed]
  135. Wei, W.; Fan, X.M.; Jia, S.H.; Zhang, X.P.; Zhang, Z.; Zhang, X.J.; Zhang, J.X.; Zhang, Y.W. Sea cucumber intestinal peptide induces the apoptosis of MCF-7 cells by inhibiting PI3K/AKT pathway. Front. Nutr. 2021, 8, 763692. [Google Scholar] [CrossRef] [PubMed]
  136. Kim, H.; Kim, H.T.; Jung, S.H.; Han, J.W.; Jo, S.; Kim, I.-G.; Kim, R.-K.; Kahm, Y.-J.; Choi, T.I.; Kim, C.H.; et al. A novel anticancerPeptide derived from bryopsisplumosa regulates proliferation and invasion in non-small cell lung cancer cells. Mar. Drugs 2023, 21, 607. [Google Scholar] [CrossRef] [PubMed]
  137. Florean, C.; Schnekenburger, M.; Lee, J.Y.; Kim, K.R.; Mazumder, A.; Song, S.; Kim, J.M.; Grandjenette, C.; Kim, J.G.; Yoon, A.Y.; et al. Discovery and characterization of isofistularin-3, a marine brominated alkaloid, as a new DNA demethylating agent inducing cell cycle arrest and sensitization to TRAIL in cancer cells. Oncotarget 2016, 7, 24027–24049. [Google Scholar] [CrossRef]
  138. Abate, A.; Tamburello, M.; Rossini, E.; Basnet, R.M.; Ribaudo, G.; Gianoncelli, A.; Hantel, C.; Cosentini, D.; Laganà, M.; Grisanti, S.; et al. Trabectedin impairs invasiveness and metastasis in adrenocortical carcinoma preclinical models. Endocr. Relat. Cancer 2022, 30, e220273. [Google Scholar] [CrossRef]
  139. Jung, U.J.; Choi, M.S. Obesity and its metabolic complications: The role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int. J. Mol. Sci. 2014, 15, 6184–6223. [Google Scholar] [CrossRef]
  140. Li, B.Y.; Xu, X.Y.; Gan, R.Y.; Sun, Q.C.; Meng, J.M.; Shang, A.; Mao, Q.Q.; Li, H.B. Targeting gut microbiota for the prevention and management of diabetes mellitus by dietary natural products. Foods 2019, 8, 440. [Google Scholar] [CrossRef]
  141. Shang, A.; Gan, R.Y.; Xu, X.Y.; Mao, Q.Q.; Zhang, P.Z.; Li, H.B. Effects and mechanisms of edible and medicinal plants on obesity: An updated review. Crit. Rev. Food Sci. Nutr. 2021, 61, 2061–2077. [Google Scholar] [CrossRef]
  142. Zang, L.; Maddison, L.A.; Chen, W. Zebrafish as a model for obesity and diabetes. Front. Cell Dev. Biol. 2018, 6, 91. [Google Scholar] [CrossRef] [PubMed]
  143. Tabassum, N.; Tai, H.; Jung, D.W.; Williams, D.R. Fishing for nature’s hits: Establishment of the zebrafish as a model for screening antidiabetic natural products. Evid. Based Complement. Altern. Med. 2015, 2015, 287847. [Google Scholar] [CrossRef]
  144. Zheng, Y.; Ley, S.H.; Hu, F.B. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat. Rev. Endocrinol. 2018, 14, 88–98. [Google Scholar] [CrossRef] [PubMed]
  145. Maddison, L.A.; Chen, W. Modeling pancreatic endocrine cell adaptation and diabetes in the zebrafish. Front. Endocrinol. 2017, 8, 9. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, X.; Han, L.W.; He, Q.X.; Han, J.; Wang, R.C. Application of zebrafish models in research of diabetes. Chin. J. Comp. Med. 2017, 27, 1–5. [Google Scholar]
  147. Lai, A.K.; Lo, A.C. Animal models of diabetic retinopathy: Summary and comparison. Diabetes Res. 2013, 2013, 106594. [Google Scholar] [CrossRef] [PubMed]
  148. Zang, L.; Shimada, Y.; Nishimura, Y.; Tanaka, T.; Nishimura, N. Repeated 14 blood collection for blood tests in adult zebrafish. J. Vis. Exp. 2015, 2015, e53272. [Google Scholar]
  149. Ilonen, J.; Lempainen, J.; Veijola, R. The heterogeneous pathogenesis of type 1 diabetes mellitus. Nat. Rev. Endocrinol. 2019, 15, 635–650. [Google Scholar] [CrossRef] [PubMed]
  150. Magkos, F.; Hjorth, M.F.; Astrup, A. Diet and exercise in the prevention and treatment of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2020, 16, 545–555. [Google Scholar] [CrossRef]
  151. Diogo, P.; Martins, G.; Simão, M.; Marreiros, A.; Eufrásio, A.C.; Cabrita, E.; Gavaia, P.J. Type I diabetes in zebrafish reduces sperm quality and increases insulin and glucose transporter transcripts. Int. J. Mol. Sci. 2023, 24, 7035. [Google Scholar] [CrossRef]
  152. Jiang, X.; Qian, H.J.; Wei, X.; Zheng, Y.X.; Zhou, Z.Y. Development and application of zebrafish diabetes model. Lab. Anim. Comp. Med. 2020, 40, 547–552. [Google Scholar]
  153. Zang, L.; Shimada, Y.; Nishimura, N. Development of a novel zebrafish model for type 2 diabetes mellitus. Sci. Rep. 2017, 7, 1461. [Google Scholar] [CrossRef]
  154. Wang, W.; Shi, Y.; Liu, Y.; Zhang, Y.; Wu, J.; Zhang, G.; Che, Q.; Zhu, T.; Li, M.; Li, D. Brasilterpenes A–E, bergamotane sesquiterpenoid derivatives with hypoglycemic activity from the deep sea-derived fungus paraconiothyrium brasiliense HDN15-135. Mar. Drugs 2022, 20, 338. [Google Scholar] [CrossRef]
  155. 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. Bioorganic Chem. 2021, 113, 104975. [Google Scholar] [CrossRef] [PubMed]
  156. Cha, S.H.; Kim, H.S.; Hwang, Y.; Jeon, Y.J.; Jun, H.S. Polysiphonia japonica extract attenuates palmitate-induced toxicity and enhances insulin secretion in pancreatic beta-cells. Oxidative Med. Cell. Longev. 2018, 2018, 4973851. [Google Scholar] [CrossRef] [PubMed]
  157. Cha, S.H.; Zhang, C.; Heo, S.J.; Jun, H.S. 5-Bromoprotocatechualdehyde combats against palmitate toxicity by inhibiting parkin degradation and reducing ros-induced mitochondrial damage in pancreatic β-cells. Antioxidants 2021, 10, 264. [Google Scholar] [CrossRef]
  158. Ahorukomeye, P.; Disotuar, M.M.; Gajewiak, J.; Karanth, S.; Watkins, M.; Robinson, S.D.; Flórez Salcedo, P.; Smith, N.A.; Smith, B.J.; Schlegel, A.; et al. Fish-hunting cone snail venoms are a rich source of minimized ligands of the vertebrate insulin receptor. eLife 2019, 8, e41574. [Google Scholar] [CrossRef] [PubMed]
  159. 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]
  160. Ji, W.; Zhang, C.H.; Allan, V.K.; Song, C. Establishment of diabetic zebrafish model and evaluation of hypoglycemic activity of antarctic krill enzymatic hydrolysates. Food Mach. 2019, 35, 24–29. [Google Scholar]
  161. Kim, E.A.; Lee, S.H.; Lee, J.H.; Kang, N.; Oh, J.Y.; Cha, S.H.; Ahn, G.; Ko, S.C.; Fernando, I.P.S.F.; Kim, S.Y. A marine algal polyphenol, dieckol, attenuates blood glucose levels by Akt pathway in alloxan induced hyperglycemia zebrafish model. RSC Adv. 2016, 6, 78570–78575. [Google Scholar] [CrossRef]
  162. Zhang, C.; Forlano, P.M.; Cone, R.D. AgRP and POMC neurons are hypophysiotropic and coordinately regulate multiple endocrine axes in a larval teleost. Cell Metab. 2012, 15, 256–264. [Google Scholar] [CrossRef] [PubMed]
  163. Seth, A.; Stemple, D.L.; Barroso, I. The emerging use of zebrafish to model metabolic disease. Dis. Models Mech. 2013, 6, 1080–1088. [Google Scholar] [CrossRef] [PubMed]
  164. Nishio, S.; Gibert, Y.; Berekelya, L.; Bernard, L.; Brunet, F.; Guillot, E.; Le Bail, J.C.; Sánchez, J.A.; Galzin, A.M.; Triqueneaux, G.; et al. Fasting induces CART down-regulation in the zebrafish nervous system in a cannabinoid receptor 1-dependent manner. Mol. Endocrinol. 2012, 26, 1316–1326. [Google Scholar] [CrossRef]
  165. Zhou, J.; Xu, Y.Q.; Guo, S.Y.; Li, C.Q. Rapid analysis of hypolipidemic drugs in a live zebrafish assay. Pharmacol. Toxicol. Methods 2015, 72, 47–52. [Google Scholar] [CrossRef] [PubMed]
  166. Oka, T.; Nishimura, Y.; Zang, L.; Hirano, M.; Shimada, Y.; Wang, Z.; Umemoto, N.; Kuroyanagi, J.; Nishimura, N.; Tanaka, T. Diet-induced obesity in zebrafish shares common pathophysiological pathways with mammalian obesity. BMC Physiol. 2010, 10, 21. [Google Scholar] [CrossRef] [PubMed]
  167. Liu, L.; Yang, Z.K.; Yang, X.T.; Yang, T.X.; Zhang, T. Construction of zebrafish obesity model and its application in lipidlowering substances. Mod. Prev. Med. 2021, 48, 2622–2625. [Google Scholar]
  168. Costa, M.; Rosa, F.; Ribeiro, T.; Hernandez-Bautista, R.; Bonaldo, M.; Gonçalves Silva, N.; Eiríksson, F.; Thorsteinsdóttir, M.; Ussar, S.; Urbatzka, R. Identification of cyanobacterial strains with potential for the treatment of obesity-related co-morbidities by bioactivity, toxicity evaluation and metabolite profiling. Mar. Drugs 2019, 17, 280. [Google Scholar] [CrossRef]
  169. Ribeiro, T.; Jónsdóttir, K.; Hernandez-Bautista, R.; Silva, N.G.; Sánchez-Astráin, B.; Samadi, A.; Eiriksson, F.F.; Thorsteinsdóttir, M.; Ussar, S.; Urbatzka, R. Metabolite profile characterization of cyanobacterial strains with bioactivity on lipid metabolism using in vivo and in vitro approaches. Mar. Drugs 2023, 21, 498. [Google Scholar] [CrossRef]
  170. Bel Mabrouk, S.; Reis, M.; Sousa, M.L.; Ribeiro, T.; Almeida, J.R.; Pereira, S.; Antunes, J.; Rosa, F.; Vasconcelos, V.; Achour, L.; et al. The marine seagrass Halophila stipulacea as a source of bioactive metabolites against obesity and biofouling. Mar. Drugs 2020, 18, 88. [Google Scholar] [CrossRef] [PubMed]
  171. Bellver, M.; Costa, S.L.d.; Sanchez, B.A.; Vasconcelos, V.; Urbatzka, R. Inhibition of intestinal lipid absorption by cyanobacterial strains in zebrafish larvae. Mar. Drugs 2021, 19, 161. [Google Scholar] [CrossRef]
  172. Luz, R.; Cordeiro, R.; Gonçalves, V.; Vasconcelos, V.; Urbatzka, R. Screening of lipid-reducing activity and cytotoxicity of the exometabolome from cyanobacteria. Mar. Drugs 2024, 22, 412. [Google Scholar] [CrossRef] [PubMed]
  173. Carrasco Del Amor, A.; Bautista, R.H.; Ussar, S.; Cristobal, S.; Urbatzka, R. Insights into the mechanism of action of the chlorophyll derivative 13-2-hydroxypheophytine a on reducing neutral lipid reserves in zebrafish larvae and mice adipocytes. Eur. J. Pharmacol. 2023, 960, 176158. [Google Scholar] [CrossRef]
  174. Silva, N.G.; Preto, M.; Vasconcelos, V.; Urbatzka, R. Reduction of neutral lipid reservoirs, bioconversion and untargeted metabolomics reveal distinct roles for vitamin K isoforms on lipid metabolism. Food Funct. 2024, 15, 2170–2180. [Google Scholar] [CrossRef] [PubMed]
  175. Santos, J.D.; Vitorino, I.; De la Cruz, M.; Díaz, C.; Cautain, B.; Annang, F.; Pérez-Moreno, G.; Gonzalez Martinez, I.; Tormo, J.R.; Martín, J.M.; et al. Bioactivities and extract dereplication of Actinomycetales isolated from marine sponges. Front. Microbiol. 2019, 10, 727. [Google Scholar] [CrossRef]
  176. Noinart, J.; Buttachon, S.; Dethoup, T.; Gales, L.; Pereira, J.A.; Urbatzka, R.; Freitas, S.; Lee, M.; Silva, A.M.S.; Pinto, M.M.M.; et al. A new ergosterol nalog, a new bis-anthraquinone and anti-obesity activity of anthraquinones froam the marine sponge-associated fungus Talaromyces stipitatus KUFA 0207. Mar. Drugs 2017, 15, 139. [Google Scholar] [CrossRef] [PubMed]
  177. Costa, M.; Coello, L.; Urbatzka, R.; Pérez, M.; Thorsteinsdottir, M. New aromatic bisabolane derivatives with lipid-reducing activity from the marine sponge Myrmekioderma sp. Mar. Drugs 2019, 17, 375. [Google Scholar] [CrossRef] [PubMed]
  178. Nakayama, H.; Shimada, Y.; Zang, L.Q.; Terasawa, M.; Nishiura, K.; Matsuda, K.; Toombs, C.; Langdon, C.; Nishimura, N. Novel anti-obesity properties of palmaria mollis in zebrafish and mouse models. Nutrients 2018, 10, 1401. [Google Scholar] [CrossRef] [PubMed]
  179. Liu, C.; Sun, Y.; Tong, X.; Xu, Z.; Xia, Q.; Xu, K. Structural characterization and hypolipidemic activity of a natural polysaccharide from Suaeda salsa L. Int. J. Biol. Macromol. 2024, 279 Pt 4, 135480. [Google Scholar] [CrossRef] [PubMed]
  180. Seo, M.J.; Seo, Y.J.; Pan, C.H.; Lee, O.H.; Kim, K.J.; Lee, B.Y. Fucoxanthin suppresses lipid accumulation and ROS production during differentiation in 3T3-L1 Adipocytes. Phytother. Res. 2016, 30, 1802–1808. [Google Scholar] [CrossRef] [PubMed]
  181. Kong, Y.; Li, Y.; Dai, Z.R.; Qin, M.; Fan, H.L.; Hao, J.G.; Zhang, C.X.; Zhong, Q.P.; Qi, C.; Wang, P. Glycosaminoglycan from Ostrea rivularis attenuates hyperlipidemia and regulates gut microbiota in high-cholesterol diet-fed zebrafish. Food Sci. Nutr. 2021, 9, 5198–5210. [Google Scholar] [CrossRef]
  182. Zhou, D.Y.; Fu, Y.H.; Li, F.T.; Yang, D.; Wei, L.N.; Yue, H.; Dai, Y.L.; Jeon, Y.J. Treatment of obese zebrafish with saringosterol acetate through AMP activated protein kinase pathway. Chem. Biodivers. 2022, 19, e202200495. [Google Scholar] [CrossRef] [PubMed]
  183. Arulselvan, P.; Fard, M.T.; Tan, W.S.; Tan, W.S.; Gothai, S.; Fakurazi, S.; Norhaizan, M.E.; Kumar, S.S. Role of antioxidants and natural products in inflammation. Oxidative Med. Cell. Longev. 2016, 2016, 5276130. [Google Scholar] [CrossRef] [PubMed]
  184. Lu, F.; Lan, Z.; Xin, Z.; He, C.; Guo, Z.; Xia, X.; Hu, T. Emerging insights into molecular mechanisms underlying pyroptosis and functions of inflammasomes in diseases. J. Cell. Physiol. 2020, 235, 3207–3221. [Google Scholar] [CrossRef] [PubMed]
  185. Li, F.L.; Zhang, Y.; Li, P.H.; Li, H.; Yang, G.W.; Liu, K.C. Anti-inflammatory activity of viridicatol based on zebrafish model. Chin. J. Antibiot. 2023, 48, 172–178. [Google Scholar]
  186. Duan, X.Y.; Ma, R.J.; Zhang, Y.; Liu, K.C. Zebrafish inflammation model and its application in anti-inflammatory field of traditional chinese medicine. Drug Eval. Res. 2021, 44, 1573–1580. [Google Scholar]
  187. Zapata, A.; Diez, B.; Cejalvo, T.; Gutiérrez-de Frías, C.; Cortés, A. Ontogeny of the immune system of fish. Fish Shellfish Immunol. 2006, 20, 126–136. [Google Scholar] [CrossRef] [PubMed]
  188. LeBert, D.C.; Huttenlocher, A. Inflammation and wound repair. Semin. Immunol. 2014, 26, 315–320. [Google Scholar] [CrossRef]
  189. Brugman, S. The zebrafish as a model to study intestinal inflammation. Dev. Comp. Immunol. 2016, 64, 82–92. [Google Scholar] [CrossRef]
  190. Cao, Y.; Xu, F.; Xia, Q.; Liu, K.; Lin, H.; Zhang, S.; Zhang, Y. The peptide LLTRAGL derived from rapana venosa exerts protective effect against inflammatory bowel disease in zebrafish model by regulating multi-pathways. Mar. Drugs 2024, 22, 100. [Google Scholar] [CrossRef] [PubMed]
  191. Gui, Y.H.; Jiao, W.H.; Zhou, M.; Zhang, Y.; Zeng, D.Q.; Zhu, H.R.; Liu, K.C.; Sun, F.; Chen, H.F.; Lin, H.W. Septosones A–C, in vivo anti-inflammatory meroterpenoids with rearranged carbon skeletons from the marine sponge dysidea septosa. Org. Lett. 2019, 21, 767–770. [Google Scholar] [CrossRef] [PubMed]
  192. Yang, F.; Sang, M.; Lu, J.R.; Zhao, H.M.; Zou, Y.; Wu, W.; Yu, Y.; Liu, Y.W.; Ma, W.; Zhang, Y.; et al. Somalactams A–D: Anti-inflammatory macrolide lactams with unique ring systems from an arctic actinomycete strain. Angew. Chem. Int. Ed. Engl. 2023, 62, e202218085. [Google Scholar] [CrossRef]
  193. Gui, Y.H.; Liu, L.; Wu, W.; Zhang, Y.; Jia, Z.L.; Shi, Y.P.; Kong, H.T.; Liu, K.C.; Jiao, W.H.; Lin, H.W. Discovery of nitrogenous sesquiterpene quinone derivatives from sponge dysidea septosa with anti-inflammatory activity in vivo zebrafish model. Bioorganic Chem. 2020, 94, 103435. [Google Scholar] [CrossRef]
  194. Li, L.; Huang, J.; Feng, L.; Xu, L.; Lin, H.; Liu, K.; Li, X.; Wang, R. Altechromone A Ameliorates Inflammatory Bowel Disease by Inhibiting NF-κB and NLRP3 Pathways. Mar. Drugs 2024, 22, 410. [Google Scholar] [CrossRef] [PubMed]
  195. Yu, X.; Han, X.; Mi, Y.; Cui, Y.; Fu, A.; Liu, K.; Li, X.; Tang, X.; Li, G. Anti-inflammatory and cytotoxicity nitrogenous merosesquiterpenoids from the sponge Pseudoceratina purpurea. Phytochemistry 2024, 226, 114220. [Google Scholar] [CrossRef] [PubMed]
  196. Zhang, X.M.; Li, H.N.; Wang, L.Z.; Zhang, S.S.; Wang, F.X.; Lin, H.W.; Gao, S.; Li, X.B.; Liu, K.C. Anti-inflammatory peptides and metabolomics-driven biomarkers discovery from sea cucumber protein hydrolysates. J. Food Sci. 2021, 86, 3540–3549. [Google Scholar] [CrossRef]
  197. Kim, E.A.; Kim, S.Y.; Ye, B.R.; Kim, J.; Ko, S.C.; Lee, W.W.; Kim, K.N.; Choi, I.W.; Jung, W.K.; Heo, S.J. Anti-inflammatory effect of Apo-9′-fucoxanthinone via inhibition of MAPKs and NF-kB signaling pathway in LPS-stimulated RAW 264.7 macrophages and zebrafish model. Int. Immunopharmacol. 2018, 59, 339–346. [Google Scholar] [CrossRef] [PubMed]
  198. Wijesinghe, W.A.; Kim, E.A.; Kang, M.C.; Lee, W.W.; Lee, H.S.; Vairappan, C.S.; Jeon, Y.J. Assessment of anti-inflammatory effect of 5β-hydroxypalisadin B isolated from red seaweed laurencia snackeyi in zebrafish embryo in vivo model. Environ. Toxicol. Pharmacol. 2014, 37, 110–117. [Google Scholar] [CrossRef]
  199. Wang, L.; Cui, Y.R.; Wang, K.; Fu, X.; Xu, J.; Gao, X.; Jeon, Y.J. Anti-inflammatory effect of fucoidan isolated from fermented sargassum fusiforme in in vitro and in vivo models. Int. J. Biol. Macromol. 2022, 222 Pt B, 2065–2071. [Google Scholar] [CrossRef]
  200. Ni, L.; Wang, L.; Fu, X.; Duan, D.; Jeon, Y.J.; Xu, J.; Gao, X. In vitro and in vivo anti-inflammatory activities of a fucose-rich fucoidan isolated from Saccharina japonica. Int. J. Biol. Macromol. 2020, 156, 717–729. [Google Scholar] [CrossRef] [PubMed]
  201. Jayawardena, T.U.; Fernando, I.P.S.; Lee, W.W.; Sanjeewa, K.K.A.; Kim, H.S.; Lee, D.S.; Jeon, Y.J. Isolation and purification of fucoidan fraction in turbinaria ornata from the maldives; inflammation inhibitory potential under LPS stimulated conditions in in-vitro and in-vivo models. Int. J. Biol. Macromol. 2019, 131, 614–623. [Google Scholar] [CrossRef] [PubMed]
  202. Samarakoon, K.W.; Kuruppu, A.I.; Ko, J.Y.; Lee, J.H.; Jeon, Y.J. Structural characterizationand anti-Inflammatory effects of 24-Methylcholesta-5(6), 22-Diene-3β-ol from the cultured marine diatom phaeodactylum tricornutum; attenuate inflammatory signaling pathways. Mar. Drugs 2023, 21, 231. [Google Scholar] [CrossRef] [PubMed]
  203. Wang, L.; Je, J.G.; Huang, C.; Oh, J.Y.; Fu, X.; Wang, K.; Ahn, G.; Xu, J.; Gao, X.; Jeon, Y.J. AntiInflammatory effect of sulfated polysaccharides isolated from codium fragile In vitro in RAW 264.7 macrophages and in vivo in zebrafish. Mar. Drugs 2022, 20, 391. [Google Scholar] [CrossRef] [PubMed]
  204. Wang, L.; Yang, H.-W.; Ahn, G.; Fu, X.; Xu, J.; Gao, X.; Jeon, Y.J. In vitro and in vivo anti-inflammatory effects of sulfated polysaccharides isolated from the edible brown seaweed, sargassum fulvellum. Mar. Drugs 2021, 19, 277. [Google Scholar] [CrossRef] [PubMed]
  205. Chen, X.; Ni, L.; Fu, X.; Wang, L.; Duan, D.; Huang, L.; Xu, J.; Gao, X. Molecular mechanism of anti-inflammatory activities of a novel sulfated galactofucan from saccharina japonica. Mar. Drugs 2021, 19, 430. [Google Scholar] [CrossRef] [PubMed]
  206. Kim, H.S.; Je, J.G.; An, H.; Baek, K.; Lee, J.M.; Yim, M.J.; Ko, S.C.; Kim, J.Y.; Oh, G.W.; Kang, M.C.; et al. Isolation and characterization of efficient active compounds using high-performance centrifugal partition chromatography (CPC) from anti-inflammatory activity fraction of ecklonia maxima in south africa. Mar. Drugs 2022, 20, 471. [Google Scholar] [CrossRef]
  207. Wang, Z.G.; Ying, X.G.; Gao, P.; Wang, C.L.; Wang, Y.F.; Yu, X.W.; Chen, J.; Wang, B.; Luo, H.Y. Anti-inflammatory activity of a peptide from skipjack (Katsuwonus pelamis). Mar. Drugs 2019, 17, 582. [Google Scholar] [CrossRef]
  208. Tang, H.; Ge, H.; Chen, Z.B.; Luo, X.M.; Su, F.J.; Liang, Y.B.; Li, Z.Y.; Wu, J.G.; Yang, Q.; Zeng, L.J.; et al. Micrometam C protects against oxidative stress in inflammation models in zebrafish and RAW264.7 macrophages. Mar. Drugs 2015, 13, 5593–5605. [Google Scholar] [CrossRef]
  209. Fei, T.T.; Liang, T.Y.; Cao, P.X.; Meng, X.; Wu, H. Research progress on the role of oxidative stress in cardiovascular disease in zebrafish. Chin. J. Comp. Med. 2024, 34, 172–178. [Google Scholar]
  210. Sies, H.; Belousov, V.V.; Chandel, N.S.; Davies, M.J.; Jones, D.P.; Mann, G.E.; Murphy, M.P.; Yamamoto, M.; Winterbourn, C. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat. Rev. Mol. Cell Biol. 2022, 23, 499–515. [Google Scholar] [CrossRef] [PubMed]
  211. Lee, H.G.; Kim, H.S.; Oh, J.Y.; Lee, D.S.; Yang, H.W.; Kang, M.C.; Kim, E.A.; Kang, N.; Kim, J.; Heo, S.J.; et al. Potential antioxidant properties of enzymatic hydrolysates from stichopus japonicus against hydrogen peroxide-induced oxidative stress. Antioxidants 2021, 10, 110. [Google Scholar] [CrossRef] [PubMed]
  212. Fang, L.; Miller, Y.I. Emerging applications for zebrafish as a model organism to study oxidative mechanisms and their roles in inflammation and vascular accumulation of oxidized lipids. Free Radic. Biol. Med. 2012, 53, 1411–1420. [Google Scholar] [CrossRef]
  213. Zhang, S.S.; Han, L.W.; Shi, Y.P.; Li, X.B.; Zhang, X.M.; Hou, H.R.; Lin, H.W.; Liu, K.C. Two novel multi-functional peptides from meat and visceral mass of marine snail Neptunea arthritica cumingii and their activities in vitro and in vivo. Mar. Drugs 2018, 16, 473. [Google Scholar] [CrossRef]
  214. Jiao, W.H.; Li, J.; Zhang, M.M.; Cui, J.; Gui, Y.H.; Zhang, Y.; Li, J.Y.; Liu, K.C.; Lin, H.W. Frondoplysins A and B, unprecedented terpene-alkaloid bioconjugates from Dysidea frondosa. Org. Lett. 2019, 21, 6190–6193. [Google Scholar] [CrossRef] [PubMed]
  215. Ko, J.Y.; Kim, E.A.; Lee, J.H.; Kang, M.C.; Lee, J.S.; Kim, J.S.; Jung, W.K.; Jeon, Y.J. Protective effect of aquacultured flounder fish-derived peptide against oxidative stress in zebrafish. Fish Shellfish Immunol. 2014, 36, 320–323. [Google Scholar] [CrossRef]
  216. Lee, H.G.; Nagahawatta, D.P.; Yang, F.; Jayawardhana, H.H.A.C.K.; Liyanage, N.M.; Lee, D.S.; Lee, J.M.; Yim, M.J.; Ko, S.C.; Kim, J.Y.; et al. Antioxidant potential of hydrolysate-derived seahorse (Hippocampus abdominalis) peptide: Protective effects against AAPH-induced oxidative damage in vitro and in vivo. Food Chem. 2023, 407, 135130. [Google Scholar] [CrossRef] [PubMed]
  217. Kang, M.C.; Cha, S.H.; Wijesinghe, W.A.; Kang, S.M.; Lee, S.H.; Kim, E.A.; Song, C.B.; Jeon, Y.J. Protective effect of marine algae phlorotannins against AAPH-induced oxidative stress in zebrafish embryo. Food Chem. 2013, 138, 950–955. [Google Scholar] [CrossRef]
  218. Wang, L.; Fu, X.; Hyun, J.; Xu, J.; Gao, X.; Jeon, Y.-J. In vitro and in vivo protective effects of agaro-oligosaccharides against hydrogen peroxide-stimulated oxidative stress. Polymers 2023, 15, 1612. [Google Scholar] [CrossRef] [PubMed]
  219. Li, Z.X.; Chen, J.W.; Yuan, F.; Huang, Y.Y.; Zhao, L.Y.; Li, J.; Su, H.X.; Liu, J.; Pang, J.Y.; Lin, Y.C.; et al. Xyloketal B exhibits its antioxidant activity through induction of HO-1 in vascular endothelial cells and zebrafish. Mar. Drugs 2013, 11, 504–522. [Google Scholar] [CrossRef]
  220. Kim, H.S.; Wang, L.; Fernando, I.P.S.; Je, J.G.; Ko, S.C.; Kang, M.C.; Lee, J.M.; Yim, M.J.; Jeon, Y.J.; Lee, D.S. Antioxidant efficacy of (−)-loliolide isolated from sargassum horneri against AAPH-induced oxidative damage in vero cells and zebrafish models in vivo. J. Appl. Phycol. 2020, 32, 3341–3348. [Google Scholar] [CrossRef]
  221. Kim, H.S.; Kim, S.Y.; Fernando, I.P.S.; Sanjeewa, K.K.A.; Wang, L.; Lee, S.H.; Ko, S.C.; Kang, M.C.; Jayawardena, T.U.; Jeon, Y.J. Free radical scavenging activity of the peptide from the alcalase hydrolysate of the edible aquacultural seahorse (Hippocampus abdominalis). J. Food Biochem. 2019, 43, e12833. [Google Scholar] [CrossRef]
  222. Wang, L.; Jayawardena, T.U.; Yang, H.W.; Lee, H.G.; Kang, M.C.; Sanjeewa, K.K.A.; Oh, J.Y.; Jeon, Y.J. Isolation, characterization, and antioxidant activity evaluation of a fucoidan from an enzymatic digest of the edible seaweed, Hizikia fusiforme. Antioxidants 2020, 9, 363. [Google Scholar] [CrossRef] [PubMed]
  223. Zhu, Y.; Yang, H.; Han, L.; Mervin, L.H.; Hosseini-Gerami, L.; Li, P.; Wright, P.; Trapotsi, M.-A.; Liu, K.; Fan, T.-P.; et al. In silico prediction and biological assessment of novel angiogenesis modulators from traditional Chinese medicine. Front. Pharmacol. 2023, 14, 1116081. [Google Scholar] [CrossRef] [PubMed]
  224. Qi, Y.; Zhao, X.; Liu, H.; Wang, Y.; Zhao, C.; Zhao, T.; Zhao, B.; Wang, Y. Identification of a Quality Marker (Q-Marker) of Danhong Injection by the Zebrafish Thrombosis Model. Molecules 2017, 22, 1443. [Google Scholar] [CrossRef]
Figure 1. Application of zebrafish models in bioactivity evaluations of marine natural products.
Figure 1. Application of zebrafish models in bioactivity evaluations of marine natural products.
Marinedrugs 22 00540 g001
Table 1. Comparison of commonly used animal models in drug discovery.
Table 1. Comparison of commonly used animal models in drug discovery.
OrganismNematode Fruit flyZebrafishMouse
Generation time3–4 days11–12 days3 months2 months
Adult size1–1.3 mm3–4 mm3–5 cm6–10 cm
Embryos size50 μm100 μm1–5 mmN/A
Brood size~140 eggs/day~120 eggs/day200–300 eggs/week6–12 pups/month
Growth conditionsSolid or liquid mediumSolid mediumLiquid mediumCages
Genome size~97 Mb~180 Mb~1500 Mb~3000 Mb
Homology to human (genome)>50%>60%>80%>90%
Transgenic organism Generationweeksweeksmonthsmonths
Culture in microtiter plateEggs to adultsEggs to larvaeEggs to larvaeN/A
High-throughput drug screening+++++++N/A
Whole biological complexity+++++++
Current use in drug discovery+++++++
Ease of experimental operation++++++++
+, ++, and +++, relative strength of the model in each category.
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

Liu, C.; Li, J.; Wang, D.; Liu, J.; Liu, K.; Li, P.; Zhang, Y. Recent Advances of the Zebrafish Model in the Discovery of Marine Bioactive Molecules. Mar. Drugs 2024, 22, 540. https://doi.org/10.3390/md22120540

AMA Style

Liu C, Li J, Wang D, Liu J, Liu K, Li P, Zhang Y. Recent Advances of the Zebrafish Model in the Discovery of Marine Bioactive Molecules. Marine Drugs. 2024; 22(12):540. https://doi.org/10.3390/md22120540

Chicago/Turabian Style

Liu, Changyu, Jiaxun Li, Dexu Wang, Jibin Liu, Kechun Liu, Peihai Li, and Yun Zhang. 2024. "Recent Advances of the Zebrafish Model in the Discovery of Marine Bioactive Molecules" Marine Drugs 22, no. 12: 540. https://doi.org/10.3390/md22120540

APA Style

Liu, C., Li, J., Wang, D., Liu, J., Liu, K., Li, P., & Zhang, Y. (2024). Recent Advances of the Zebrafish Model in the Discovery of Marine Bioactive Molecules. Marine Drugs, 22(12), 540. https://doi.org/10.3390/md22120540

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