Development and Application of Herbal Medicine from Marine Origin

A special issue of Marine Drugs (ISSN 1660-3397).

Deadline for manuscript submissions: closed (28 February 2019) | Viewed by 45305

Printed Edition Available!
A printed edition of this Special Issue is available here.

Special Issue Editors


E-Mail Website
Guest Editor
Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taiwan;
Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taiwan
Interests: neutrophilic inflammation; innate immunity; signal transduction; protein kinase; drug research and development
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
National Museum of Marine Biology and Aquarium, Pingtung 944401, Taiwan
Interests: marine natural products; marine chemical ecology; bioactive substances from cultured marine invertebrates
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
1. Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
2. Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
3. Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
4. Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei 110, Taiwan
Interests: marine microbial natural products, de-replication, symbiotic microbes, biofunctional activities

Special Issue Information

Dear Colleagues,

Marine herbal medicine generally refers to the use of marine plants as original materials to develop crude drugs, or for other medical use. The term ‘marine plants’ usually denotes macroalgae grown between intertidal and sub-intertidal zones, including Chlorophyta, Phaeophyta and Rhodophyta. Considerable progress has been made in the field of biomedical research into marine microalgae and microorganisms in the past decade. As the most important source of fundamental products in the world, marine plants have a very important role in biomedical research. Furthermore, worldwide studies have consistently demonstrated that many crude drugs derived from marine plants contain novel ingredients that may benefit health or can be used in the treatment of diseases; some have been developed into health foods, and some even into drugs. It is expected that there are many substances of marine plant origin that will have medical applications in terms of improving human health, and are awaiting discovery.

In this Special Issue, Development and Application of Herbal Medicine of Marine Origin, we will provide a platform for researchers to publish biomedical studies on substances of marine plant origin. We welcome submissions from scientists and academics from across the world. Selected papers from this conference "The 33rd Symposium on Natural Products" (https://nphs.kmu.edu.tw/index.php/zh-TW/nph33) will also be considered to be published in this Special Issue.

Prof. Dr. Tsong-Long Hwang
Prof. Dr. Ping-Jyun Sung
Assoc. Prof. Dr. Chih-Chuang Liaw
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Marine Drugs is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Herbal medicine

  • Algae

  • Microorganism

  • Biomedicine

  • Disease

  • Drug

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

15 pages, 7344 KiB  
Article
Phomaketide A Inhibits Lymphangiogenesis in Human Lymphatic Endothelial Cells
by Huai-Ching Tai, Tzong-Huei Lee, Chih-Hsin Tang, Lei-Po Chen, Wei-Cheng Chen, Ming-Shian Lee, Pei-Chi Chen, Chih-Yang Lin, Chih-Wen Chi, Yu-Jen Chen, Cheng-Ta Lai, Shiou-Sheng Chen, Kuang-Wen Liao, Chien-Hsing Lee and Shih-Wei Wang
Mar. Drugs 2019, 17(4), 215; https://0-doi-org.brum.beds.ac.uk/10.3390/md17040215 - 06 Apr 2019
Cited by 16 | Viewed by 4525
Abstract
Lymphangiogenesis is an important biological process associated with cancer metastasis. The development of new drugs that block lymphangiogenesis represents a promising therapeutic strategy. Marine fungus-derived compound phomaketide A, isolated from the fermented broth of Phoma sp. NTOU4195, has been reported to exhibit anti-angiogenic [...] Read more.
Lymphangiogenesis is an important biological process associated with cancer metastasis. The development of new drugs that block lymphangiogenesis represents a promising therapeutic strategy. Marine fungus-derived compound phomaketide A, isolated from the fermented broth of Phoma sp. NTOU4195, has been reported to exhibit anti-angiogenic and anti-inflammatory effects. However, its anti-lymphangiogenic activity has not been clarified to date. In this study, we showed that phomaketide A inhibited cell growth, migration, and tube formation of lymphatic endothelial cells (LECs) without an evidence of cytotoxicity. Mechanistic investigations revealed that phomaketide A reduced LECs-induced lymphangiogenesis via vascular endothelial growth factor receptor-3 (VEGFR-3), protein kinase Cδ (PKCδ), and endothelial nitric oxide synthase (eNOS) signalings. Furthermore, human proteome array analysis indicated that phomaketide A significantly enhanced the protein levels of various protease inhibitors, including cystatin A, serpin B6, tissue factor pathway inhibitor (TFPI), and tissue inhibitor matrix metalloproteinase 1 (TIMP-1). Importantly, phomaketide A impeded tumor growth and lymphangiogenesis by decreasing the expression of LYVE-1, a specific marker for lymphatic vessels, in tumor xenograft animal model. These results suggest that phomaketide A may impair lymphangiogenesis by suppressing VEGFR-3, PKCδ, and eNOS signaling cascades, while simultaneously activating protease inhibitors in human LECs. We document for the first time that phomaketide A inhibits lymphangiogenesis both in vitro and in vivo, which suggests that this natural product could potentially treat cancer metastasis. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Graphical abstract

8 pages, 1256 KiB  
Article
Components from the Leaves and Twigs of Mangrove Lumnitzera racemosa with Anti-Angiogenic and Anti-Inflammatory Effects
by Szu-Yin Yu, Shih-Wei Wang, Tsong-Long Hwang, Bai-Luh Wei, Chien-Jung Su, Fang-Rong Chang and Yuan-Bin Cheng
Mar. Drugs 2018, 16(11), 404; https://0-doi-org.brum.beds.ac.uk/10.3390/md16110404 - 25 Oct 2018
Cited by 8 | Viewed by 4205
Abstract
One new neolignan, racelactone A (1), together with seven known compounds (28) were isolated from the methanolic extract of the leaves and twigs of Lumnitzera racemosa. The structure of racelactone A (1) was determined [...] Read more.
One new neolignan, racelactone A (1), together with seven known compounds (28) were isolated from the methanolic extract of the leaves and twigs of Lumnitzera racemosa. The structure of racelactone A (1) was determined on the basis of the mass and NMR spectroscopic data interpretation. With respect to bioactivity, compound 1 displayed an anti-angiogenic effect by suppressing tube formation. Furthermore, compounds 1, 4, and 5 showed significant anti-inflammatory effects with IC50 values of 4.95 ± 0.89, 1.95 ± 0.40, and 2.57 ± 0.23 μM, respectively. The plausible biosynthesis pathway of racelactone A (1) was proposed. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Figure 1

14 pages, 8316 KiB  
Article
Fucoidan Inhibits Radiation-Induced Pneumonitis and Lung Fibrosis by Reducing Inflammatory Cytokine Expression in Lung Tissues
by Hsin-Hsien Yu, Edward Chengchuan KO, Chia-Lun Chang, Kevin Sheng-Po Yuan, Alexander T. H. Wu, Yan-Shen Shan and Szu-Yuan Wu
Mar. Drugs 2018, 16(10), 392; https://0-doi-org.brum.beds.ac.uk/10.3390/md16100392 - 19 Oct 2018
Cited by 46 | Viewed by 7382
Abstract
Purpose: Radiotherapy is a crucial treatment approach for many types of cancer. Radiation pneumonitis (RP) is one of the major complications in chest irradiation. Fucoidan is a sulfated polysaccharide found mainly in various species of brown seaweed. Recent studies have demonstrated the [...] Read more.
Purpose: Radiotherapy is a crucial treatment approach for many types of cancer. Radiation pneumonitis (RP) is one of the major complications in chest irradiation. Fucoidan is a sulfated polysaccharide found mainly in various species of brown seaweed. Recent studies have demonstrated the anti-inflammatory effects of fucoidan. However, no study has reported a well-established prophylactic agent for RP. Therefore, we investigated the effects of fucoidan on RP and radiotherapy (RT)-induced lung fibrosis. Materials and Methods: We compared RP and RT-induced fibrosis in lung tissue specimens obtained from irradiated (10 Gy/shot) C57BL/6 mice with or without fucoidan administration (200 mg/kg/day, oral gavage for 14 days). The expression patterns of cytokines in the pleural fluid were determined using a cytokine array and confirmed through enzyme immunoassays. Results: Fucoidan administration attenuated RP and RT-induced fibrosis in lung tissues. Decreased neutrophil and macrophage accumulation was observed in irradiated lung tissues, and radiation-induced lung fibrosis, as demonstrated by Masson trichrome staining, was attenuated. We investigated the expression patterns of inflammatory cytokines in the irradiated lung pleural fluid through the protein array; results revealed that fucoidan administration changed the expression patterns of inflammatory cytokines in irradiated lung tissues. Furthermore, the expression levels of TIMP-1, CXCL1, MCP-1, MIP-2, and interleukin-1Ra were substantially enhanced in the pleural fluid, but fucoidan administration significantly reduced their expression. Conclusions: Fucoidan changes the expression patterns of inflammatory cytokines, which may consequently attenuate RP and RT-induced lung fibrosis. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Graphical abstract

16 pages, 2836 KiB  
Article
The Protective Role of Sulfated Polysaccharides from Green Seaweed Udotea flabellum in Cells Exposed to Oxidative Damage
by Fernando Bastos Presa, Maxsuell Lucas Mendes Marques, Rony Lucas Silva Viana, Leonardo Thiago Duarte Barreto Nobre, Leandro Silva Costa and Hugo Alexandre Oliveira Rocha
Mar. Drugs 2018, 16(4), 135; https://0-doi-org.brum.beds.ac.uk/10.3390/md16040135 - 20 Apr 2018
Cited by 37 | Viewed by 4234
Abstract
Seaweed is a rich source of bioactive sulfated polysaccharides. We obtained six sulfated polysaccharide-rich fractions (UF-0.3, UF-0.5, UF-0.6, UF-0.7, UF-1.0, and UF-2.0) from the green seaweed Udotea flabellum (UF) by proteolytic digestion followed by sequential acetone precipitation. Biochemical analysis of these fractions showed [...] Read more.
Seaweed is a rich source of bioactive sulfated polysaccharides. We obtained six sulfated polysaccharide-rich fractions (UF-0.3, UF-0.5, UF-0.6, UF-0.7, UF-1.0, and UF-2.0) from the green seaweed Udotea flabellum (UF) by proteolytic digestion followed by sequential acetone precipitation. Biochemical analysis of these fractions showed that they were enriched with sulfated galactans. The viability and proliferative capacity of 3T3 fibroblasts exposed to FeSO4 (2 µM), CuSO4 (1 µM) or ascorbate (2 mM) was not affected. However, these cells were exposed to oxidative stress in the presence of FeSO4 or CuSO4 and ascorbate, which caused the activation of caspase-3 and caspase-9, resulting in apoptosis of the cells. We also observed increased lipid peroxidation, evaluated by the detection of malondialdehyde and decreased glutathione and superoxide dismutase levels. Treating the cells with the ultrafiltrate fractions (UF) fractions protected the cells from the oxidative damage caused by the two salts and ascorbate. The most effective protection against the oxidative damage caused by iron was provided by UF-0.7 (1.0 mg/mL); on treatment with UF-0.7, cell viability was 55%. In the case of copper, cell viability on treatment with UF-0.7 was ~80%, but the most effective fraction in this model was UF-2.0, with cell viability of more than 90%. The fractions, mainly UF-0.7 and UF-2.0, showed low iron chelating activity, but high copper chelating activity and total antioxidant capacity (TAC). These results suggested that some of their protective mechanisms stem from these properties. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Figure 1

13 pages, 3590 KiB  
Article
The Bioactive Extract of Pinnigorgia sp. Induces Apoptosis of Hepatic Stellate Cells via ROS-ERK/JNK-Caspase-3 Signaling
by Liang-Mou Kuo, Po-Jen Chen, Ping-Jyun Sung, Yu-Chia Chang, Chun-Ting Ho, Yi-Hsiu Wu and Tsong-Long Hwang
Mar. Drugs 2018, 16(1), 19; https://0-doi-org.brum.beds.ac.uk/10.3390/md16010019 - 09 Jan 2018
Cited by 40 | Viewed by 5380
Abstract
The activation of hepatic stellate cells (HSCs) is a significant phenomenon during the pathogenesis of liver disorders, including liver cirrhosis and fibrosis. Here, we identified that the extract from a gorgonian coral Pinnigorgia sp. (Pin) induced apoptosis of HSC-T6 cells. Pin inhibited the [...] Read more.
The activation of hepatic stellate cells (HSCs) is a significant phenomenon during the pathogenesis of liver disorders, including liver cirrhosis and fibrosis. Here, we identified that the extract from a gorgonian coral Pinnigorgia sp. (Pin) induced apoptosis of HSC-T6 cells. Pin inhibited the viability of HSC-T6 cells and increased their subG1 population, DNA fragmentation, caspase-3 activation, and reactive oxygen species (ROS) production in a concentration-dependent manner. The Pin-induced ROS generation and apoptotic effects were significantly reversed by a thiol antioxidant, N-acetylcysteine (NAC). Additionally, Pin induced ERK/JNK phosphorylation and pharmacological inhibition of ERK/JNK rescued the Pin-induced cell death. Pin-activated ERK/JNK were significantly reduced after the administration of NAC; however, the inhibition of ERK/JNK failed to change the Pin-induced ROS production. Similarly, pinnigorgiol A, a pure compound isolated from Pin, elicited ROS production and apoptosis in HSC-T6 cells. The pinnigorgiol A-induced apoptosis was retrained by NAC. Together, it appears that Pin leads to apoptosis in HSC-T6 cells through ROS-mediated ERK/JNK signaling and caspase-3 activation. Pinnigorgiol A serves as a bioactive compound of Pin and may exhibit therapeutic potential by clearance of HSCs. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Graphical abstract

1067 KiB  
Article
Angiotensin I-Converting Enzyme (ACE) Inhibitory Activity, Antioxidant Properties, Phenolic Content and Amino Acid Profiles of Fucus spiralis L. Protein Hydrolysate Fractions
by Lisete Paiva, Elisabete Lima, Ana Isabel Neto and José Baptista
Mar. Drugs 2017, 15(10), 311; https://0-doi-org.brum.beds.ac.uk/10.3390/md15100311 - 13 Oct 2017
Cited by 71 | Viewed by 4890
Abstract
Food protein-derived hydrolysates with multi-bioactivities such as antihypertensive and antioxidant properties have recently received special attention since both activities can play significant roles in preventing cardiovascular diseases. This study reports, for the first time, the angiotensin I-converting enzyme (ACE)-inhibition and antioxidant properties of [...] Read more.
Food protein-derived hydrolysates with multi-bioactivities such as antihypertensive and antioxidant properties have recently received special attention since both activities can play significant roles in preventing cardiovascular diseases. This study reports, for the first time, the angiotensin I-converting enzyme (ACE)-inhibition and antioxidant properties of ultrafiltrate fractions (UF) with different molecular weight ranges (<1, 1–3 and ≥3 kDa) obtained from Fucus spiralis protein hydrolysate (FSPH) digested with cellulase–bromelain. The amino acids profile, recovery yield, protein, peptide and total phenolic contents of these FSPH-UF, and the in vitro digestibility of F. spiralis crude protein were also investigated. FSPH-UF ≥3 kDa presented remarkably higher ACE-inhibition, yield, peptide and polyphenolic (phlorotannins) contents. Antioxidant analysis showed that FSPH-UF <1 kDa and ≥3 kDa exhibited significantly higher scavenging of 2,2-diphenyl-1-picrylhydrazyl radical and ferrous ion-chelating (FIC) activity. FSPH-UF ≥3 kDa had also notably higher ferric reducing antioxidant power (FRAP). Strong correlations were observed between ACE-inhibition and antioxidant activities (FIC and FRAP). The results suggest that ACE-inhibition and antioxidant properties of FSPH-UF may be due to the bioactive peptides and polyphenols released during the enzymatic hydrolysis. In conclusion, this study shows the potential use of defined size FSPH-UF for the prevention/treatment of hypertension and/or oxidative stress-related diseases. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Figure 1

1981 KiB  
Article
Pharmacokinetics of Jaspine B and Enhancement of Intestinal Absorption of Jaspine B in the Presence of Bile Acid in Rats
by Min-Koo Choi, Jihoon Lee, So Jeong Nam, Yun Ju Kang, Youjin Han, Kwangik Choi, Young A. Choi, Mihwa Kwon, Dongjoo Lee and Im-Sook Song
Mar. Drugs 2017, 15(9), 279; https://0-doi-org.brum.beds.ac.uk/10.3390/md15090279 - 01 Sep 2017
Cited by 7 | Viewed by 3745
Abstract
We aimed to investigate the pharmacokinetics and the underlying mechanisms of the intestinal absorption, distribution, metabolism, and excretion of Jaspine B in rats. The oral bioavailability of Jaspine B was 6.2%, but it decreased to 1.6% in bile-depleted rats and increased to 41.2% [...] Read more.
We aimed to investigate the pharmacokinetics and the underlying mechanisms of the intestinal absorption, distribution, metabolism, and excretion of Jaspine B in rats. The oral bioavailability of Jaspine B was 6.2%, but it decreased to 1.6% in bile-depleted rats and increased to 41.2% (normal) and 23.5% (bile-depleted) with taurocholate supplementation (60 mg/kg). Consistent with the increased absorption in the presence of bile salts, rat intestinal permeability of Jaspine B also increased in the presence of 10 mM taurocholate or 20% bile. Further studies demonstrated that the enhanced intestinal permeability with bile salts was due to increased lipophilicity and decreased membrane integrity. Jaspine B was designated as a highly tissue-distributed compound, because it showed large tissue to plasma ratios in the brain, kidney, heart, and spleen. Moreover, the recovery of Jaspine B from the feces and urine after an intravenous administration was about 6.3%, suggesting a substantial metabolism of Jaspine B. Consistent with this observation, 80% of the administered Jaspine B was degraded after 1 h incubation with rat liver microsomes. In conclusion, the facilitated intestinal permeability in the presence of bile salts could significantly increase the bioavailability of Jaspine B and could lead to the development of oral formulations of Jaspine B with bile salts. Moreover, the highly distributed features of Jaspine B in the brain, kidney, heart, and spleen should be carefully considered in the therapeutic effect and toxicity of this compound. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Graphical abstract

3465 KiB  
Article
Inhibitory Growth of Oral Squamous Cell Carcinoma Cancer via Bacterial Prodigiosin
by Ming-Fang Cheng, Chun-Shu Lin, Yu-Hsin Chen, Ping-Jyun Sung, Shian-Ren Lin, Yi-Wen Tong and Ching-Feng Weng
Mar. Drugs 2017, 15(7), 224; https://0-doi-org.brum.beds.ac.uk/10.3390/md15070224 - 15 Jul 2017
Cited by 36 | Viewed by 5091
Abstract
Chemotherapy drugs for oral cancers always cause side effects and adverse effects. Currently natural sources and herbs are being searched for treated human oral squamous carcinoma cells (OSCC) in an effort to alleviate the causations of agents in oral cancers chemotherapy. This study [...] Read more.
Chemotherapy drugs for oral cancers always cause side effects and adverse effects. Currently natural sources and herbs are being searched for treated human oral squamous carcinoma cells (OSCC) in an effort to alleviate the causations of agents in oral cancers chemotherapy. This study investigates the effect of prodigiosin (PG), an alkaloid and natural red pigment as a secondary metabolite of Serratia marcescens, to inhibit human oral squamous carcinoma cell growth; thereby, developing a new drug for the treatment of oral cancer. In vitro cultured human OSCC models (OECM1 and SAS cell lines) were used to test the inhibitory growth of PG via cell cytotoxic effects (MTT assay), cell cycle analysis, and Western blotting. PG under various concentrations and time courses were shown to effectively cause cell death and cell-cycle arrest in OECM1 and SAS cells. Additionally, PG induced autophagic cell death in OECM1 and SAS cells by LC3-mediated P62/LC3-I/LC3-II pathway at the in vitro level. These findings elucidate the role of PG, which may target the autophagic cell death pathways as a potential agent in cancer therapeutics. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Graphical abstract

Review

Jump to: Research

3573 KiB  
Review
Terpenoids from Octocorals of the Genus Pachyclavularia
by Yu-Chia Chang, Jyh-Horng Sheu, Yang-Chang Wu and Ping-Jyun Sung
Mar. Drugs 2017, 15(12), 382; https://0-doi-org.brum.beds.ac.uk/10.3390/md15120382 - 05 Dec 2017
Cited by 12 | Viewed by 4040
Abstract
In this paper, we reviewed natural compounds isolated from octocorals belonging to the genus Pachyclavularia, including 20 cembrane-, 39 briarane-, and eight briarellin-related diterpenoids, and one secosterol. The chemical constituents of these 68 secondary metabolites, and their names, structures, and bioactivities, along [...] Read more.
In this paper, we reviewed natural compounds isolated from octocorals belonging to the genus Pachyclavularia, including 20 cembrane-, 39 briarane-, and eight briarellin-related diterpenoids, and one secosterol. The chemical constituents of these 68 secondary metabolites, and their names, structures, and bioactivities, along with studies of their biological activities, are summarized in this review. Based on the literature, many of these compounds possess bioactivities, including anti-inflammation properties, cytotoxicity, and ichthyotoxicity, suggesting that they may have the potential to be developed into biomedical agents for treatment. Full article
(This article belongs to the Special Issue Development and Application of Herbal Medicine from Marine Origin)
Show Figures

Figure 1

Back to TopTop