1
|
Liang BE, Long LS, Wu XY, Huang MY, Lai Y, Yuan X, Wang MH, Li M, Zheng QQ, Zhang HL, Chen MC, Liu ZD, Geng X, Lyu QQ, Wang WD, Liu QH, Liu WZ, Li CL. Alginate oligosaccharide prevents renal ischemia-reperfusion injury in rats via MRC1-mediated pathway. Acta Pharmacol Sin 2025:10.1038/s41401-025-01545-3. [PMID: 40263568 DOI: 10.1038/s41401-025-01545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 04/24/2025]
Abstract
Acute kidney injury (AKI) is a clinical syndrome that is defined as a sudden decline in renal function and characterized by inflammation and tubular injury. Alginate oligosaccharide (AOSC), a natural product obtained from alginate by acidolysis and hydrolysis, shows activities of antioxidant, immunomodulation, and anti-inflammation. In this study, we investigated the potential of AOSC in the treatment of AKI. Renal ischemia-reperfusion (I/R) was induced in male rats by clipping both the renal artery and vein for 45 min followed by reperfusion for 24 h. The rats were treated with AOSC (100 mg/kg, i.g.) before surgery. At the end of the experiments, both kidneys were collected for protein, mRNA measurement, or histological analysis. We showed that AOSC pretreatment significantly improved glomerular and tubular function in the kidney of I/R rats. AOSC markedly inhibited I/R-induced activation of TLR4/MyD88/NF-κB/IL-1β inflammatory signaling and prevented apoptosis in the kidney. In HK2 cells subjected to hypoxia/reoxygenation (H/R) stimulation, AOSC (250-1000 μg/ml) dose-dependently prevented pro-inflammatory responses and cell apoptosis. Transcriptomic analysis revealed that I/R increased the expression levels of mannose receptor type C1 (MRC1) in the kidney, which was markedly inhibited by AOSC. Molecular docking showed that AOSC interacted with E725, N727, E733, T743, S745, and N747 of MRC1 through hydrogen bonds. MRC1 gene knockout significantly improved renal function and attenuated I/R-induced kidney inflammation and apoptosis in mice. In line with this, AOSC failed to prevent I/R-induced kidney injury in MRC1 gene knockout mice. UPLC analysis showed that the protection of AOSC in HK2 cells subjected to H/R was likely attributed to MRC1-mediated intracellular endocytosis. In conclusion, AOSC prevents I/R-induced AKI, which is at least partially mediated by MRC1.
Collapse
Affiliation(s)
- Bai-En Liang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Luo-Sha Long
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin-Yan Wu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mei-Ying Huang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying Lai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xi Yuan
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ming-Hui Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qi-Qi Zheng
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hai-Ling Zhang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Man-Chun Chen
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhen-de Liu
- Haitang (Jiangsu) Biotechnology Co Ltd, Nantong, 226100, China
| | - Xin Geng
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Qian-Qian Lyu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Wei-Dong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Hua Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
- Department of Nephrology, Jieyang People's Hospital, Jieyang, 522000, China.
| | - Wei-Zhi Liu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Chun-Ling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Tu L, Xing B, Ma S, Zou Z, Wang S, Feng J, Cheng M, Jin Y. A review on polysaccharide-based tumor targeted drug nanodelivery systems. Int J Biol Macromol 2025; 304:140820. [PMID: 39933669 DOI: 10.1016/j.ijbiomac.2025.140820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
The tumor-targeted drug delivery system (TTDNS) uses nanocarriers to transport chemotherapeutic agents to target tumor cells or tissues precisely. This innovative approach considerably increases the effective concentration of these drugs at the tumor site, thereby enhancing their therapeutic efficacy. Many chemotherapeutic agents face challenges, such as low bioavailability, high cytotoxicity, and inadequate drug resistance. To address these obstacles, TTDNS comprising natural polysaccharides have gained increasing popularity in the field of nanotechnology owing to their ability to improve safety, bioavailability, and biocompatibility while reducing toxicity. In addition, it enhances permeability and allows for controlled drug delivery and release. This review focuses on the sources of natural polysaccharides and their direct and indirect mechanisms of anti-tumor activity. We also explored the preparation of various polysaccharide-based nanocarriers, including nanoparticles, nanoemulsions, nanohydrogels, nanoliposomes, nanocapsules, nanomicelles, nanocrystals, and nanofibers. Furthermore, this review delves into the versatile applications of polysaccharide-based nanocarriers, elucidating their capabilities for in vivo targeting, controlled release, and responsiveness to endogenous and exogenous stimuli, such as pH, reactive oxygen species, glutathione, light, ultrasound, and magnetic fields. This sophisticated design substantially enhances the chemotherapeutic efficacy of the encapsulated drugs at tumor sites and provides a basis for preclinical and clinical research. However, the in vivo stability, drug loading, and permeability of these preparations into tumor tissues still need to be improved. Most of the currently developed biomarker-sensitive polysaccharide nanocarriers are still in the laboratory stage, more innovative delivery mechanisms and clinical studies are needed to develop commercial nanocarriers for medical use.
Collapse
Affiliation(s)
- Liangxing Tu
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Banghuai Xing
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Shufei Ma
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Zijian Zou
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Siying Wang
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Jianfang Feng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China; Guangxi University of Chinese Medicine, Nanning 530200, PR China.
| | - Meng Cheng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| | - Yi Jin
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| |
Collapse
|
3
|
Pradhan B, Ki JS. Seaweed-derived laminarin and alginate as potential chemotherapeutical agents: An updated comprehensive review considering cancer treatment. Int J Biol Macromol 2025; 293:136593. [PMID: 39426775 DOI: 10.1016/j.ijbiomac.2024.136593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
Seaweed-derived bioactive substances such as polysaccharides have proven to be effective chemotherapeutic and chemopreventive agents. Laminarin and alginate antioxidant properties aid in the prevention of cancer through dynamic modulation of critical intracellular signaling pathways via apoptosis which produce low cytotoxicity and potential chemotherapeutic effects. Understanding the effects of laminarin and alginate on human cancer cells and their molecular roles in cell death pathways can help to develop a novel chemoprevention strategy. This review emphasizes the importance of apoptosis-modulating laminarin and alginate in a range of malignancies as well as their extraction, molecular structure, and weight. In addition, future nano-formulation enhancements for greater clinical efficacy are discussed. Laminarin and alginate are perfect ingredients because of their distinct physicochemical and biological characteristics and their use-based delivery systems in cancer. The effectiveness of laminarin and alginate against cancer and more preclinical and clinical trials will open up as new chemotherapeutic natural drugs which lead to established as potential cancer drugs.
Collapse
Affiliation(s)
- Biswajita Pradhan
- Department of Life Science, Sangmyung University, Seoul 03016, South Korea; Department of Botany, Model Degree College, Rayagada 765017, Odisha, India
| | - Jang-Seu Ki
- Department of Life Science, Sangmyung University, Seoul 03016, South Korea.
| |
Collapse
|
4
|
Kareem RA, Razavi SH, Mousavi ZE. Effect of Sodium Alginate-Bulk Chitosan/Chitosan Nanoparticle Wall Matrix on the Viability of Lactobacillus plantarum Under Simulated Gastrointestinal Fluids. Appl Biochem Biotechnol 2025; 197:1991-2011. [PMID: 39630335 DOI: 10.1007/s12010-024-05105-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
The viability of probiotic cells decreases during passage through the gastrointestinal tract. The process of probiotics encapsulation with sodium alginate and chitosan polymers was carried out to protect the Lactobacillus plantarum in adverse conditions. Lactobacillus plantarum was entrapped in sodium alginate/chitosan (SA/BChi) and sodium alginate/nano-chitosan (SA/NChi) wall materials. Encapsulating L. plantarum with SA/BChi and SA/NChi resulted in a high encapsulation efficiency % of ~ 86.41 to 91.09%. In addition, coating bacteria cells in encapsulants improved the survivability of the cells under the simulated gastrointestinal fluids by ~ 52.61% in SA/Chi and 58.04% in SA/NChi compared to 29% for unencapsulated forms. Probiotic beads under field emission-scanning electron microscopy (FE-SEM) were morphologically compact with a cracked appearance of SA/NChi beads. The Fourier transform-infrared spectroscopy (FT-IR) and differential scanning calorimetry (DSC) showed vigorous electrostatic interaction between polymers, as well as the high melting points, which corroborate the previous investigations in the field for using SA/BChi or SA/NChi as a promising encapsulating agent for ameliorating the survivability of probiotics under harsh conditions. The distinctive properties possessed by the two coatings make them excellent candidates for use as polymeric carriers in probiotic delivery systems.
Collapse
Affiliation(s)
- Raghda Abdulhussain Kareem
- Directorate of Agricultural Extension and Training, Ministry of Agriculture, Basrah, Iraq
- Department of Food Science, Engineering and Technology, Faculty of Agricultural Engineering and Technology, University of Tehran, Karaj, Iran
| | - Seyed Hadi Razavi
- Department of Food Science, Engineering and Technology, Faculty of Agricultural Engineering and Technology, University of Tehran, Karaj, Iran.
| | - Zeinab E Mousavi
- Department of Food Science, Engineering and Technology, Faculty of Agricultural Engineering and Technology, University of Tehran, Karaj, Iran
| |
Collapse
|
5
|
Ye Y, Zhang Y, Zhou Y, Gao Y. Molecular Engineering of Alginate Lyases and the Potential Agricultural Applications of Their Enzymatic Products. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40011194 DOI: 10.1021/acs.jafc.4c09913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Alginate lyases, enzymes that degrade alginate into unsaturated oligosaccharides, have attracted significant attention for their potential applications across various fields, particularly in agriculture. This review focuses on the molecular engineering of alginate lyases to enhance their activity, stability, and specificity as well as the agricultural applications of the resulting enzymatic products, known as alginate oligosaccharides (AOS). We start by summarizing the sources and classification of alginate lyases, followed by recent advances in their engineering through directed evolution, rational design, truncation of noncatalytic domains, and conserved domain reconstruction. We then explore the diverse agricultural applications of AOS, including their ability to promote plant growth, to increase the content of active plant components, to extend fruit shelf life, and to enhance plant resistance to abiotic stresses. Furthermore, the potential value of AOS as feed additives and preservatives in shrimp-based products is also assessed. This review will not only lay a solid theoretical foundation but also serve as a catalyst for the innovative development and practical application of high-value enzymatic preparations and utilization of AOS-related products, providing new solutions for sustainable agriculture and the food industry.
Collapse
Affiliation(s)
- Yongshang Ye
- Department of Biotechnology, School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China
| | - Yu Zhang
- Department of Biotechnology, School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China
| | - Ying Zhou
- Department of Biotechnology, School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China
| | - Yuhang Gao
- Department of Biotechnology, School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China
| |
Collapse
|
6
|
He Z, Meng S, Xu Y, Zhong M, Han X, Xie Q, Ding M, Li J, Hu Z. Direct Influence of the Conserved Motif in PL7 Family Alginate Lyases on Enzyme Cold Adaptability. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4320-4330. [PMID: 39932157 DOI: 10.1021/acs.jafc.4c10895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Alginate lyase, a vital component of polysaccharide lyases, is instrumental in the efficient degradation of alginate and the production of single oligosaccharides. Although numerous alginate lyases have been characterized, only a few display extreme cold adaptability in the range of 0-20 °C. In this study, we identified a novel cold-adapted alginate lyase, Aly423, from Tamlana laminarinivorans PT2-4 isolated from Sargassum. Phylogenetic classification, enzyme structure, and catalytic property analyses confirmed that Aly423 could be classified as a member of subfamily 5 of the PL7 family and exhibited significant cold adaptability at low temperatures. Further analysis of the secondary structure and homology modeling of several cold-adapted enzymes revealed two variable amino acid sites in the conserved amino acid motif (YFK*G*Y) of Aly423, which may affect the cold adaptation mechanism. Point mutation experiments demonstrated that mutant A304T significantly altered the temperature adaptation of Aly423, highlighting the critical role of this amino acid site in the cold-adaptation mechanism of the enzyme. In summary, we effectively enhanced the enzymatic activity of the PL7 alginate cold-adapted enzyme through a rational design using computational methods. This advancement is of significant importance for the efficient utilization of sodium alginate in the food, agricultural, and pharmaceutical industries under low-temperature conditions.
Collapse
Affiliation(s)
- Zhixiao He
- Department of Biology, College of Science, Shantou University, Shantou 515063, China
| | - Shanshan Meng
- Department of Biology, College of Science, Shantou University, Shantou 515063, China
| | - Yan Xu
- Department of Biology, Heyuan Polytechnic, Heyuan 517000, China
| | - Mingqi Zhong
- Department of Biology, College of Science, Shantou University, Shantou 515063, China
| | - Xuefeng Han
- Department of Biology, College of Science, Shantou University, Shantou 515063, China
| | - Qingyi Xie
- Haikou Key Laboratory for Research and Utilization of Tropical Natural Products, Institute of Tropical Bioscience and Biotechnology, CATAS, Haikou 571101, China
| | - Mo Ding
- Department of Biology, College of Science, Shantou University, Shantou 515063, China
| | - Jin Li
- Guangdong Provincial Key Laboratory of Marine Disaster Prediction and Prevention, Shantou University, Shantou 515063, China
- College of Life Sciences, China West Normal University, Nanchong 637002, China
| | - Zhong Hu
- Department of Biology, College of Science, Shantou University, Shantou 515063, China
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou 515063, China
| |
Collapse
|
7
|
Torlopov M, Shevchenko O, Drozd N, Udoratina E. Ethylenediamine-modified alginate - A hemocompatible platform for polymer-drug conjugates. Int J Biol Macromol 2025; 287:138326. [PMID: 39638197 DOI: 10.1016/j.ijbiomac.2024.138326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
The study is dedicated to the synthesis, rheological properties, hemocompatibility, and further modification of water-soluble derivatives of sodium alginate containing fragments of ethylenediamine (Alg-EDA). Alg-EDA with an equal ratio of amide/amine groups and varying degrees of substitution were synthesized by the carbodiimide method. The influence of the molecular weight of Alg-EDA on the attachment of bioactive molecules such as hydroxybenzoic and ferulic acids was determined. Modification of alginate with ethylenediamine fragments leads to a reduction in dynamic viscosity and sensitivity to Ca2+ ions (internal gelation method). Alg-EDA derivatives, with differ in molecular weight and degree of substitution with phenolic acids, are characterized by high hemocompatibility (in vitro tests: erythrocyte hemolysis, blood recalcification time, activated partial thromboplastin time). Antioxidant properties of the synthesized alginate derivatives were characterized using models of varying complexity (including cellular models). It was found that Alg-EDA, at a concentration of 0.5 mg/mL, had a statistically significant membrane-protective activity under conditions of acute oxidative stress induced by both H2O2 and AAPH. Derivatives with lower molecular weight were more effective than high molecular weight ones. Modification of polysaccharides by the fragments of phenolic acids into the structure contributed to the enhancement of antioxidant properties. The hemocompatible macromolecular antioxidants synthesized in this study are promising for further in-depth investigation for the creation of biomedical materials.
Collapse
Affiliation(s)
- Mikhail Torlopov
- Institute of Chemistry of Komi Scientific Centre of the Ural Branch of the Russian Academy of Sciences, Pervomayskaya st. 48, Syktyvkar 167000, Komi Republic, Russian Federation
| | - Oksana Shevchenko
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the Russian Academy of Sciences, Kommunisticheskaya st. 28, Syktyvkar 167982, Komi Republic, Russian Federation
| | - Natalia Drozd
- National Medical Research Center for Hematology, Novy Zykovsky passage 4, Moscow 125167, Russian Federation
| | - Elena Udoratina
- Institute of Chemistry of Komi Scientific Centre of the Ural Branch of the Russian Academy of Sciences, Pervomayskaya st. 48, Syktyvkar 167000, Komi Republic, Russian Federation.
| |
Collapse
|
8
|
Li J, Shao M, Liu H, Guo P, Liu F, Ma M, Li Q. Lithium Coupled with C6-Carboxyl Improves the Efficacy of Oligoguluronate in DSS-Induced Ulcerative Colitis in C57BL/6J Mice. Mar Drugs 2024; 22:573. [PMID: 39728147 DOI: 10.3390/md22120573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Abstract
Oligoguluronate lithium (OGLi) was prepared for the purpose of enhancing the anti-ulcerative colitis (UC) activities of OG, in which lithium (Li+) is coupled with the C6-carboxyl of G residue. The therapeutic effects of OGLi on dextran sulfate (DSS)-induced UC mice were investigated, and oligoguluronate sodium (OGNa) and lithium carbonate (LC) were used as contrasts. The effects of OGLi, OGNa and LC on the treatment of UC mice were studied by monitoring body weight change and evaluating colon length, the disease activity index (DAI), histopathological examination and gut microbiota regulation. The results showed that compared with OGNa and LC, OGLi significantly reduced the clinical symptoms and histopathological changes associated with UC in the acute model. It was worth noting that OGLi significantly changed the gut microbiota characteristics of the DSS-treated mice and corrected the typical dysbacteriosis of DSS-induced UC. This intervention resulted in increasing the abundance of norank_f_Muribaculaceae and Ileibacterium spp. while reducing the levels of Escherichia-Shigella spp. and Romboutsia spp. The OGLi could significantly increase the diversity of intestinal microorganisms in the short term. All of these discoveries demonstrate that lithium collaboratively enhances the anti-UC efficacy of OG, which will help to create OG-based drugs for the treatment of UC.
Collapse
Affiliation(s)
- Jiayi Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Meng Shao
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Hao Liu
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Peng Guo
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Fei Liu
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Mingfeng Ma
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Quancai Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| |
Collapse
|
9
|
Grobler CE, Mabate B, Prins A, Le Roes-Hill M, Pletschke BI. Expression, Purification, and Characterisation of Recombinant Alginate Lyase ( Flammeovirga AL2) for the Bioconversion of Alginate into Alginate Oligosaccharides. Molecules 2024; 29:5578. [PMID: 39683737 DOI: 10.3390/molecules29235578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Alginate, a polysaccharide found in brown seaweeds, has regularly gained attention for its potential use as a source of bioactive compounds. However, it is structurally complex with a high molecular weight, limiting its application. Alginate oligosaccharides (AOS) are small, soluble fragments, making them more bioavailable. Alginate hydrolysis by enzymes is the preferred method for AOS production. Commercially available alginate lyases are limited, expensive, and sometimes exhibit unsatisfactory activity, making the search for novel alginate lyases with improved activity indispensable. The aims of this study were to codon-optimise, synthesise, express, purify, and characterise a recombinant alginate lyase, AL2, from Flammeovirga sp. strain MY04 and to compare it to a commercial alginate lyase. Expression was successfully performed using Escherichia coli ArcticExpress (DE3) RP cells, and the protein was purified through affinity chromatography. The recombinant enzyme was characterised by pH optimum studies, and temperature optimum and stability experiments. The optimal reaction conditions for AL2 were pH 9.0 and 37 °C, while for the commercial enzyme, the optimal conditions were pH 8.0 and 37 °C. At optimal reaction conditions, the specific activity of AL2 was 151.6 ± 12.8 µmol h-1 mg-1 protein and 96.9 ± 13.1 µmol h-1 mg-1 protein for the commercial alginate lyase. Moreover, AL2 displayed impressive activity in breaking down alginate into AOS. Hence, AL2 shows potential for use as an industrial enzyme for the hydrolysis of alginate into alginate oligosaccharides. Additional studies should be carried out to further characterise this enzyme, improve its purity, and optimise its activity.
Collapse
Affiliation(s)
- Coleen E Grobler
- Enzyme Science Programme (ESP), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6140, South Africa
| | - Blessing Mabate
- Enzyme Science Programme (ESP), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6140, South Africa
| | - Alaric Prins
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa
| | - Marilize Le Roes-Hill
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa
| | - Brett I Pletschke
- Enzyme Science Programme (ESP), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6140, South Africa
| |
Collapse
|
10
|
Mi J, Tong Y, Zhang Q, Wang Q, Wang Y, Wang Y, Lin G, Ma Q, Li T, Huang S. Alginate Oligosaccharides Enhance Gut Microbiota and Intestinal Barrier Function, Alleviating Host Damage Induced by Deoxynivalenol in Mice. J Nutr 2024; 154:3190-3202. [PMID: 39357672 DOI: 10.1016/j.tjnut.2024.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 09/17/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Alginate oligosaccharides (AOS) exhibits notable effects in terms of anti-inflammatory, antibacterial, and antioxidant properties. Deoxynivalenol (DON) has the potential to trigger intestinal inflammation by upregulating pro-inflammatory cytokines and apoptosis, thereby compromising the integrity of the intestinal barrier function and perturbing the balance of the gut microbiota. OBJECTIVES We assessed the impact of AOS on mitigating DON-induced intestinal damage and systemic inflammation in mice. METHODS After a 1-wk acclimatization period, the mice were divided into 4 groups. For 3 wk, the AOS and AOS + DON groups were gavaged daily with 200 μL of AOS [200 mg/kg body weight (BW)], whereas the CON and DON groups received an equivalent volume of sterile Phosphate-Buffered Saline (PBS). Subsequently, for 1 wk, the DON and AOS + DON groups received 100 μL of DON (4.8 mg/kg BW) daily, whereas the control (CON) and AOS groups continued receiving PBS. RESULTS After administering DON via gavage to mice, there was a significant decrease (P < 0.05) in body weights compared with the CON group. Interestingly, AOS exhibited a tendency to mitigate this weight loss in the AOS + DON group. In the feces of mice treated with both AOS and DON, the concentration of DON significantly increased (P < 0.05) compared with the DON group alone. Histological analysis revealed that DON exposure caused increased intestinal damage, including shortened villi and eroded epithelial cells, which was ameliorated by presupplementation with AOS, alleviating harm to the intestinal barrier function. In both jejunum and colon tissues, DON exposure significantly reduced (P < 0.05) the expression of tight junction proteins (claudin and occludin in the colon) and the mucin protein mucin 2, compared with the CON group. Prophylactic administration of AOS alleviated these reductions, thereby improving the expression levels of these key proteins. Additionally, AOS supplementation protected DON-exposed mice by increasing the abundance of probiotics such as Bifidobacterium, Faecalibaculum, and Romboutsia. These gut microbes are known to enhance (P < 0.05) anti-inflammatory responses and the production of short-chain fatty acids (SCFAs), including total SCFAs, acetate, and valerate, compared with the DON group. CONCLUSIONS This study unveils that AOS not only enhances gut microbiota and intestinal barrier function but also significantly mitigates DON-induced intestinal damage.
Collapse
Affiliation(s)
- Jinqiu Mi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China; Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Yaoyi Tong
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China; Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Qiyue Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China; Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China; College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Qingfeng Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China; Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Yanwei Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China; Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China; School of Life Science, Shanxi University, Taiyuan, Shanxi, China
| | - Yue Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China; Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Gang Lin
- Institute of Quality Standards and Testing Technology for Agricultural Products, Chinese Academy of Agricultural Science, Beijing, China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China; Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China
| | - Tiantian Li
- Institute of Cereal & Oil Science and Technology, Academy of National Food and Strategic Reserves Administration, Beijing, China.
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China; Feed Safety and Healthy Livestock, Beijing Jingwa Agricultural Innovation Center, Beijing, China.
| |
Collapse
|
11
|
Lukova P, Kokova V, Baldzhieva A, Murdjeva M, Katsarov P, Delattre C, Apostolova E. Alginate from Ericaria crinita Possesses Antioxidant Activity and Attenuates Systemic Inflammation via Downregulation of Pro-Inflammatory Cytokines. Mar Drugs 2024; 22:482. [PMID: 39590762 PMCID: PMC11595431 DOI: 10.3390/md22110482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Alginates are anionic polysaccharides present in the cell walls of brown seaweeds. Various biological activities of alginate and its derivatives have been described. In this study, we assessed the potential of alginate obtained from Ericaria crinita (formerly Cystoseira crinita) to scavenge free radicals and function as a ferric ion reductor. The anti-inflammatory effect on the serum levels of TNF-α, IL-1β, IL-6, and IL-10 of rats with LPS-induced systemic inflammation after 14 days of treatment was also examined. Ericaria crinita alginate showed antioxidant activities of IC50 = 505 µg/mL (DPPH) and OD700 > 2 (ferric reducing power). A significant decrease in serum levels of IL-1β was observed only in animals treated with the polysaccharide at a dose of 100 mg/kg bw. Both doses of E. crinita alginate (25 and 100 mg/kg bw) significantly reduced the serum concentrations of pro-inflammatory cytokines TNF-α and IL-6, but no statistical significance was observed in the levels of the anti-inflammatory cytokine IL-10. Our findings show the potential of E. crinita alginate to act as an antioxidant and anti-inflammatory agent. It is likely that the exhibited antioxidant ability of the polysaccharide contributes to its antiphlogistic effects. More in-depth studies are needed to fully understand the specific mechanisms and the molecular pathways involved in these activities.
Collapse
Affiliation(s)
- Paolina Lukova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
| | - Vesela Kokova
- Department of Pharmacology, Toxicology, and Pharmacotherapy, Faculty of Pharmacy, Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
| | - Alexandra Baldzhieva
- Department of Medical Microbiology and Immunology “Prof. Dr. Elissay Yanev”, Faculty of Medicine, Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
- Research Institute at Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
| | - Marianna Murdjeva
- Department of Medical Microbiology and Immunology “Prof. Dr. Elissay Yanev”, Faculty of Medicine, Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
- Research Institute at Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
| | - Plamen Katsarov
- Research Institute at Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
| | - Cédric Delattre
- Clermont Auvergne INP, CNRS, Institut Pascal, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- Institut Universitaire de France (IUF), 1 rue Descartes, 75005 Paris, France
| | - Elisaveta Apostolova
- Department of Pharmacology, Toxicology, and Pharmacotherapy, Faculty of Pharmacy, Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
- Research Institute at Medical University-Plovdiv, Vasil Aprilov Str. 15A, 4002 Plovdiv, Bulgaria
| |
Collapse
|
12
|
Wang H, Wen J, Ablimit N, Deng K, Wang W, Jiang W. Degradation of Natural Undaria pinnatifida into Unsaturated Guluronic Acid Oligosaccharides by a Single Alginate Lyase. Mar Drugs 2024; 22:453. [PMID: 39452861 PMCID: PMC11509462 DOI: 10.3390/md22100453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Here, we report on a bifunctional alginate lyase (Vnalg7) expressed in Pichia pastoris, which can degrade natural Undaria pinnatifida into unsaturated guluronic acid di- and trisaccharide without pretreatment. The enzyme activity of Vnalg7 (3620.00 U/mL-culture) was 15.81-fold higher than that of the original alg (228.90 U/mL-culture), following engineering modification. The degradation rate reached 52.75%, and reducing sugar reached 30.30 mg/mL after combining Vnalg7 (200.00 U/mL-culture) and 14% (w/v) U. pinnatifida for 6 h. Analysis of the action mode indicated that Vnalg7 could degrade many substrates to produce a variety of unsaturated alginate oligosaccharides (AOSs), and the minimal substrate was tetrasaccharide. Site-directed mutagenesis showed that Glu238, Glu241, Glu312, Arg236, His307, Lys414, and Tyr418 are essential catalytic sites, while Glu334, Glu344, and Asp311 play auxiliary roles. Mechanism analysis revealed the enzymatic degradation pattern of Vnalg7, which mainly recognizes and attacks the third glycosidic linkage from the reducing end of oligosaccharide substrate. Our findings provide a novel alginate lyase tool and a sustainable and commercial production strategy for value-added biomolecules using seaweeds.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Jiang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, Beijing 100193, China; (H.W.); (J.W.); (N.A.); (K.D.); (W.W.)
| |
Collapse
|
13
|
Li QQ, Xu D, Dong QW, Song XJ, Chen YB, Cui YL. Biomedical potentials of alginate via physical, chemical, and biological modifications. Int J Biol Macromol 2024; 277:134409. [PMID: 39097042 DOI: 10.1016/j.ijbiomac.2024.134409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Alginate is a linear polysaccharide with a modifiable structure and abundant functional groups, offers immense potential for tailoring diverse alginate-based materials to meet the demands of biomedical applications. Given the advancements in modification techniques, it is significant to analyze and summarize the modification of alginate by physical, chemical and biological methods. These approaches provide plentiful information on the preparation, characterization and application of alginate-based materials. Physical modification generally involves blending and physical crosslinking, while chemical modification relies on chemical reactions, mainly including acylation, sulfation, phosphorylation, carbodiimide coupling, nucleophilic substitution, graft copolymerization, terminal modification, and degradation. Chemical modified alginate contains chemically crosslinked alginate, grafted alginate and oligo-alginate. Biological modification associated with various enzymes to realize the hydrolysis or grafting. These diverse modifications hold great promise in fully harnessing the potential of alginate for its burgeoning biomedical applications in the future. In summary, this review provides a comprehensive discussion and summary of different modification methods applied to improve the properties of alginate while expanding its biomedical potentials.
Collapse
Affiliation(s)
- Qiao-Qiao Li
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Dong Xu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Qin-Wei Dong
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Xu-Jiao Song
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Yi-Bing Chen
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
14
|
Zhou Y, Wei Z, Tan J, Sun H, Jiang H, Gao Y, Zhang H, Schroyen M. Alginate oligosaccharide supplementation improves boar semen quality under heat stress. STRESS BIOLOGY 2024; 4:37. [PMID: 39251532 PMCID: PMC11383898 DOI: 10.1007/s44154-024-00177-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/23/2024] [Indexed: 09/11/2024]
Abstract
Heat stress is a serious problem that affects animal husbandry by reducing growth and reproductive performance of animals. Adding plant extracts to the diet is an effective way to help overcome this problem. Alginate oligosaccharide (AOS) is a natural non-toxic antioxidant with multiple biological activities. This study analyzed the potential mechanism of AOS in alleviating heat stress and improving semen quality in boars through a combination of multiple omics tools. The results indicated that AOS could significantly increase sperm motility (P < 0.001) and sperm concentration (P < 0.05). At the same time, AOS improved the antioxidant capacity of blood and semen, and increased blood testosterone (P < 0.05) level. AOS could improve the metabolites in sperm, change the composition of gut microbiota, increase the relative abundance of beneficial bacteria such as Pseudomonas (P < 0.01), Escherichia-Shigella (P < 0.05), Bifidobacterium (P < 0.01), reduce the relative abundance of harmful bacteria such as Prevotella_9 (P < 0.05), Prevotellaceae_UCG-001 (P < 0.01), and increase the content of short chain fatty acids. Proteomic results showed that AOS increased proteins related to spermatogenesis, while decreasing heat shock protein 70 (P < 0.05) and heat shock protein 90 (P < 0.01). These results were verified using immunofluorescence staining technology. There was a good correlation among sperm quality, sperm metabolome, sperm proteome, and gut microbiota. In conclusion, AOS can be used as a feed additive to increase the semen quality of boars to enhance reproductive performance under heat stress.
Collapse
Affiliation(s)
- Yexun Zhou
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, P. R. China
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Zeou Wei
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, P. R. China
- School of Agriculture and Food Science, University College Dublin, Belfeld, Dublin 4, Ireland
| | - Jiajian Tan
- YangXiang Joint Stock Company, Guigang, 53700, China
| | - Haiqing Sun
- YangXiang Joint Stock Company, Guigang, 53700, China
| | - Haidi Jiang
- YangXiang Joint Stock Company, Guigang, 53700, China
| | - Yang Gao
- College of Life Science, Baicheng Normal University, Baicheng, 137000, Jilin, China.
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, P. R. China.
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| |
Collapse
|
15
|
Huang Q, Wu W, Wen Y, Lu S, Zhao C. Potential therapeutic natural compounds for the treatment of Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155822. [PMID: 38909512 DOI: 10.1016/j.phymed.2024.155822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a complicated neurodegenerative disease with cognitive impairment occurring in the older people, in which extracellular accumulation of β-amyloid and intracellular aggregation of hyperphosphorylated tau are regarded as the prevailing theories. However, the exact AD mechanism has not been determined. Moreover, there is no effective treatment available in phase III trials to eradicate AD, which is imperative to explore novel treatments. PURPOSE A number of up-to-date pre-clinical studies on cognitive impairment is beneficial to clarify the pathology of AD. This review recapitulates several advances in AD pathobiology and discusses the neuroprotective effects of natural compounds, such as phenolic compounds, natural polysaccharides and oligosaccharides, peptide, and lipids, underscoring the therapeutic potential for AD. METHODS Electronic databases involving PubMed, Web of Science, and Google Scholar were searched up to October 2023. Articles were conducted using the keywords like Alzheimer's disease, pathogenic mechanisms, natural compounds, and neuroprotection. RESULT This review summarized several AD pathologies and the neuroprotective effects of natural compounds such as natural polysaccharides and oligosaccharides, peptide, and lipids. CONCLUSION We have discussed the pathogenic mechanisms of AD and the effect natural products on neurodegenerative diseases particularly in treating AD. Specifically, we investigated the molecular pathways and links between natural compounds and Alzheimer's disease such as through NF-κB, Nrf2, and mTOR pathway. Further investigation is necessary in exploring the bioactivity and effectiveness of natural compounds in clinical trials, which may provide a promising treatment for AD patients.
Collapse
Affiliation(s)
- Qihui Huang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, 36310 Vigo, Spain
| | - Weihao Wu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yuxi Wen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, 36310 Vigo, Spain
| | - Suyue Lu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chao Zhao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
16
|
Wu X, Xin Y, Zhang H, Quan L, Ao Q. Biopolymer-Based Nanomedicine for Cancer Therapy: Opportunities and Challenges. Int J Nanomedicine 2024; 19:7415-7471. [PMID: 39071502 PMCID: PMC11278852 DOI: 10.2147/ijn.s460047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/18/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer, as the foremost challenge among human diseases, has plagued medical professionals for many years. While there have been numerous treatment approaches in clinical practice, they often cause additional harm to patients. The emergence of nanotechnology has brought new directions for cancer treatment, which can deliver anticancer drugs specifically to tumor areas. This article first introduces the application scenarios of nanotherapies and treatment strategies of nanomedicine. Then, the noteworthy characteristics exhibited by biopolymer materials were described, which make biopolymers stand out in polymeric nanomedicine delivery. Next, we focus on summarizing the state-of-art studies of five categories of proteins (Albumin, Gelatin, Silk fibroin, Zein, Ferritin), nine varieties of polysaccharides (Chitosan, Starch, Hyaluronic acid, Dextran, cellulose, Fucoidan, Carrageenan, Lignin, Pectin) and liposomes in the field of anticancer drug delivery. Finally, we also provide a summary of the advantages and limitations of these biopolymers, discuss the prevailing impediments to their application, and discuss in detail the prospective research directions. This review not only helps readers understand the current development status of nano anticancer drug delivery systems based on biopolymers, but also is helpful for readers to understand the properties of various biopolymers and find suitable solutions in this field through comparative reading.
Collapse
Affiliation(s)
- Xixi Wu
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Yuan Xin
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Hengtong Zhang
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Liang Quan
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Qiang Ao
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| |
Collapse
|
17
|
Chen Y, Chen B, Dong J, Yang D, Tang H, Wen L, Li J, Huang L, Zhou J. A tough and bioadhesive injectable hydrogel formed with maleimidyl alginate and pristine gelatin. Carbohydr Polym 2024; 334:122011. [PMID: 38553212 DOI: 10.1016/j.carbpol.2024.122011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/01/2024] [Accepted: 02/29/2024] [Indexed: 04/02/2024]
Abstract
Injectable hydrogels have wide applications in clinical practice. However, the development of tough and bioadhesive ones based on biopolymers, along with biofriendly and robust crosslinking strategies, still represents a great challenge. Herein, we report an injectable hydrogel composed of maleimidyl alginate and pristine gelatin, for which the precursor solutions could self-crosslink via mild Michael-type addition without any catalyst or external energy upon mixing. This hydrogel is tough and bioadhesive, which can maintain intactness as well as adherence to the defect of porcine skin under fierce bending and twisting, warm water bath, and boiling water shower. Besides, it is biocompatible, bioactive and biodegradable, which could support the growth and remodeling of cells by affording an extracellular matrix-like environment. As a proof of application, we demonstrate that this hydrogel could significantly accelerate diabetic skin wound healing, thereby holding great potential in healthcare.
Collapse
Affiliation(s)
- Yin Chen
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China; School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Baiqi Chen
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jianpei Dong
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Deyu Yang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Hao Tang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Lan Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianshu Li
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China; College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Lu Huang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jianhua Zhou
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
18
|
Acevedo S, Covarrubias AA, Haeger P, Pancetti F, Tala F, de la Fuente-Ortega E. Alginate Oligosaccharides Protect Gastric Epithelial Cells against Oxidative Stress Damage through Induction of the Nrf2 Pathway. Antioxidants (Basel) 2024; 13:618. [PMID: 38790723 PMCID: PMC11117588 DOI: 10.3390/antiox13050618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/01/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Gastric diseases represent a significant global public health challenge, characterized by molecular dysregulation in redox homeostasis and heightened oxidative stress. Although prior preclinical studies have demonstrated the cytoprotective antioxidant effects of alginate oligosaccharides (AOSs) through the Nrf2 pathway, whether such mechanisms apply to gastric diseases remains unclear. In this study, we used the GES-1 gastric cell line exposed to hydrogen peroxide (H2O2) as a damage model to investigate the impact of AOS on cell viability and its associated mechanisms. Our results revealed that pre-incubation with AOS for either 4 h or 24 h significantly improved the viability of GES-1 cells exposed to H2O2. In addition, AOS reduced the intracellular ROS levels, activating the Nrf2 signaling pathway, with increased Nrf2 protein and mRNA expression and a significant upregulation of the target genes HO-1 and NQO1. The activation of Nrf2 was correlated with decreased Keap1 protein expression and an increased level of the autophagy protein p62/SQSTM1, suggesting the activation of Nrf2 through a noncanonical pathway. This study suggests that AOS is a potential treatment for protecting gastric epithelial cells from oxidative stress by activating the p62/SQSTM1-Keap1-Nrf2 axis and laying the foundation for future investigations about its specific therapeutic mechanisms.
Collapse
Affiliation(s)
- Samantha Acevedo
- Laboratorio de Estrés Celular y Enfermedades Crónicas no Transmisibles, Universidad Católica del Norte, Coquimbo 1781421, Chile;
| | - Alejandra A. Covarrubias
- Laboratorio de Neurotoxicología Ambiental, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo 1781421, Chile
- Facultad de Ciencias Agropecuarias, Universidad del Alba, La Serena 1700000, Chile
| | - Paola Haeger
- Laboratorio de Neurobiología de la Conducta, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo 1781421, Chile;
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago 8370186, Chile
- Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Universidad Católica del Norte, Coquimbo 1781421, Chile;
| | - Floria Pancetti
- Laboratorio de Neurotoxicología Ambiental, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo 1781421, Chile
- Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Universidad Católica del Norte, Coquimbo 1781421, Chile;
- Centro de Investigación y Desarrollo Tecnológico en Algas y Otros Recursos Biológicos (CIDTA), Facultad de Ciencias del Mar, Universidad Católica del Norte, Coquimbo 1781421, Chile
| | - Fadia Tala
- Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Universidad Católica del Norte, Coquimbo 1781421, Chile;
- Centro de Investigación y Desarrollo Tecnológico en Algas y Otros Recursos Biológicos (CIDTA), Facultad de Ciencias del Mar, Universidad Católica del Norte, Coquimbo 1781421, Chile
- Departamento de Biología Marina, Facultad de Ciencias del Mar, Universidad Católica del Norte, Coquimbo 1781421, Chile
- Instituto Milenio en Socio-Ecología Costera, SECOS, Santiago 7550000, Chile
| | - Erwin de la Fuente-Ortega
- Laboratorio de Estrés Celular y Enfermedades Crónicas no Transmisibles, Universidad Católica del Norte, Coquimbo 1781421, Chile;
- Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Universidad Católica del Norte, Coquimbo 1781421, Chile;
- Centro de Investigación y Desarrollo Tecnológico en Algas y Otros Recursos Biológicos (CIDTA), Facultad de Ciencias del Mar, Universidad Católica del Norte, Coquimbo 1781421, Chile
| |
Collapse
|
19
|
Mohammed V, Arockiaraj J. Unveiling the trifecta of cyanobacterial quorum sensing: LuxI, LuxR and LuxS as the intricate machinery for harmful algal bloom formation in freshwater ecosystems. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 924:171644. [PMID: 38471587 DOI: 10.1016/j.scitotenv.2024.171644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 02/22/2024] [Accepted: 03/09/2024] [Indexed: 03/14/2024]
Abstract
Harmful algal blooms (HABs) are causing significant disruptions in freshwater ecosystems, primarily due to the proliferation of cyanobacteria. These blooms have a widespread impact on various lakes globally, leading to profound environmental and health consequences. Cyanobacteria, with their ability to produce diverse toxins, pose a particular concern as they negatively affect the well-being of humans and animals, exacerbating the situation. Notably, cyanobacteria utilize quorum sensing (QS) as a complex communication mechanism that facilitates coordinated growth and toxin production. QS plays a critical role in regulating the dynamics of HABs. However, recent advances in control and mitigation strategies have shown promising results in effectively managing and reducing the occurrence of HABs. This comprehensive review explores the intricate aspects of cyanobacteria development in freshwater ecosystems, explicitly focusing on deciphering the signaling molecules associated with QS and their corresponding genes. Furthermore, a concise overview of diverse measures implemented to efficiently control and mitigate the spread of these bacteria will be provided, shedding light on the ongoing global efforts to address this urgent environmental issue. By deepening our understanding of the mechanisms driving cyanobacteria growth and developing targeted control strategies, we hope to safeguard freshwater ecosystems and protect the health of humans and animals from the detrimental impacts of HABs.
Collapse
Affiliation(s)
- Vajagathali Mohammed
- Department of Forensic Science, Yenepoya Institute of Arts, Science, Commerce, and Management, Yenepoya (Deemed to be University), Mangaluru 575013, Karnataka, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
20
|
Sharma S, Kishen A. Bioarchitectural Design of Bioactive Biopolymers: Structure-Function Paradigm for Diabetic Wound Healing. Biomimetics (Basel) 2024; 9:275. [PMID: 38786486 PMCID: PMC11117869 DOI: 10.3390/biomimetics9050275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Chronic wounds such as diabetic ulcers are a major complication in diabetes caused by hyperglycemia, prolonged inflammation, high oxidative stress, and bacterial bioburden. Bioactive biopolymers have been found to have a biological response in wound tissue microenvironments and are used for developing advanced tissue engineering strategies to enhance wound healing. These biopolymers possess innate bioactivity and are biodegradable, with favourable mechanical properties. However, their bioactivity is highly dependent on their structural properties, which need to be carefully considered while developing wound healing strategies. Biopolymers such as alginate, chitosan, hyaluronic acid, and collagen have previously been used in wound healing solutions but the modulation of structural/physico-chemical properties for differential bioactivity have not been the prime focus. Factors such as molecular weight, degree of polymerization, amino acid sequences, and hierarchical structures can have a spectrum of immunomodulatory, anti-bacterial, and anti-oxidant properties that could determine the fate of the wound. The current narrative review addresses the structure-function relationship in bioactive biopolymers for promoting healing in chronic wounds with emphasis on diabetic ulcers. This review highlights the need for characterization of the biopolymers under research while designing biomaterials to maximize the inherent bioactive potency for better tissue regeneration outcomes, especially in the context of diabetic ulcers.
Collapse
Affiliation(s)
- Shivam Sharma
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON M5G 1G6, Canada
| | - Anil Kishen
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON M5G 1G6, Canada
- Department of Dentistry, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| |
Collapse
|
21
|
Bajpai D, G K. Development and Evaluation of Alginate- and Carrageenan-Incorporated Scaffold for Bone Regeneration: An In Vitro Study. Cureus 2024; 16:e61139. [PMID: 38933614 PMCID: PMC11200317 DOI: 10.7759/cureus.61139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Periodontitis, a persistent inflammatory condition, results in the deterioration of both the hard and soft tissues in the periodontium, leading to the formation of intrabony defects. Restoring the lost tissues, particularly bone, is possible through tissue engineering techniques utilizing scaffolds made from different polymers. Consequently, this research focuses on creating and assessing a scaffold infused with alginate (Sigma Aldrich, Gillingham, UK) and carrageenan (Sigma Aldrich, Gillingham, UK) for the purpose of bone regeneration. METHODS An in vitro investigation was conducted to assess the characteristics of the recently formulated scaffold. Spectroscopic analysis, tensile strength testing, scanning electron microscopy (SEM) analysis, and degradation testing were carried out to evaluate both the physical and biological attributes of the scaffold. RESULTS IBM SPSS Statistics for Windows, V. 1.2 (IBM Corp., Armonk, NY, USA) was used for statistical analysis. A one-way ANOVA test was done to determine the significance of tensile strength, and a paired t-test was done to check the significance of the degradation test. The in vitro research unveiled notable distinctions in the physical and biological attributes between the scaffold infused with alginate and carrageenan and the PerioCol® (p<0.05). CONCLUSION The scaffold incorporating alginate and carrageenan demonstrated superior outcomes concerning parameters such as tensile stress and strain, degradation rate, percentage bone volume, and object surface density when contrasted with the conventional PerioCol®. Therefore, the scaffold infused with alginate and carrageenan emerges as a promising candidate for bone regeneration.
Collapse
Affiliation(s)
- Devika Bajpai
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Kaarthikeyan G
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
22
|
Huang MY, An YC, Zhang SY, Qiu SJ, Yang YY, Liu WC. Metabolomic analysis reveals biogenic selenium nanoparticles improve the meat quality of thigh muscle in heat-stressed broilers is related to the regulation of ferroptosis pathway. Poult Sci 2024; 103:103554. [PMID: 38401225 PMCID: PMC10906527 DOI: 10.1016/j.psj.2024.103554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 02/26/2024] Open
Abstract
Heat stress (HS) causes oxidative damage and abnormal metabolism of muscle, thus impairing the meat quality in broilers. Selenium is an indispensable element for enhancing antioxidant systems. In our previous study, we synthesized a novel type of biogenic selenium nanoparticles synthesized with alginate oligosaccharides (SeNPs-AOS), and found that the particle size of Se is 80 nm and the Se content is 8% in the SeNPs-AOS; and dietary 5 mg/kg SeNPs-AOS has been shown to be effective against HS in broilers. However, whether SeNPs-AOS can mitigate HS-induced the impairment of thigh muscle quality in broilers is still unclear. Therefore, the purpose of this study was to investigate the protective effects of dietary SeNPs-AOS on meat quality, antioxidant capacity, and metabolomics of thigh muscle in broilers under HS. A total of 192 twenty-one-day-old Arbor Acres broilers were randomly divided into 4 groups with 6 replicates per group (8 broilers per replicate) according to a 2 × 2 experimental design: thermoneutral group (TN, broilers raised under 23±1.5°C); TN+SeNPs-AOS group (TN group supplemented 5 mg/kg SeNPS-AOS); HS group (broilers raised under 33 ± 2°C for 10 h/d); and HS + SeNPs-AOS group (HS group supplemented 5 mg/kg SeNPS-AOS). The results showed that HS increased the freezing loss, cooking loss, and malondialdehyde (MDA) content of thigh muscle, whereas decreased the total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) activities, as well as downregulated the mRNA expression of SOD2, CAT, GPX3, nuclear factor erythroid 2-related factor 2 (Nrf2), selenoprotein S (SELENOS), solute carrier family 7 member 11 (SLC7A11), GPX4, and ferroportin 1 (Fpn1) of thigh muscle (P < 0.05). Dietary SeNPS-AOS reduced the b* value, elevated the pH0min value and the activities of T-SOD, GSH-Px, glutathione S-transferase (GST) and the mRNA expression levels of GSTT1, GSTA3, GPX1, GPX3, ferritin heavy polypeptide-1 (FTH1), and Fpn1 of thigh muscle in broilers under HS (P < 0.05). Nontargeted metabolomics analysis identified a total of 79 metabolites with significant differences among the four groups, and the differential metabolites were mainly enriched in 8 metabolic pathways including glutathione metabolism and ferroptosis (P < 0.05). In summary, dietary 5 mg/kg SeNPs-AOS (Se content of 8%) could alleviate HS-induced impairment of meat quality by improving the oxidative damage, metabolic disorders and ferroptosis of thigh muscle in broilers challenged with HS. Suggesting that the SeNPs-AOS may be used as a novel nano-modifier for meat quality in broilers raised in thermal environment.
Collapse
Affiliation(s)
- Meng-Yi Huang
- Department of Animal Science, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yu-Chen An
- Yangjiang Campus of Guangdong Ocean University, Yangjiang, 529500, China
| | - Shu-Yue Zhang
- Department of Animal Science, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang 524088, China
| | - Sheng-Jian Qiu
- Department of Animal Science, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yu-Ying Yang
- Department of Animal Science, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang 524088, China
| | - Wen-Chao Liu
- Department of Animal Science, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
23
|
Chen C, Li X, Lu C, Zhou X, Chen L, Qiu C, Jin Z, Long J. Advances in alginate lyases and the potential application of enzymatic prepared alginate oligosaccharides: A mini review. Int J Biol Macromol 2024; 260:129506. [PMID: 38244735 DOI: 10.1016/j.ijbiomac.2024.129506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Alginate is mainly a linear polysaccharide composed of randomly arranged β-D-mannuronic acid and α-L-guluronic acid linked by α, β-(1,4)-glycosidic bonds. Alginate lyases degrade alginate mainly adopting a β-elimination mechanism, breaking the glycosidic bonds between the monomers and forming a double bond between the C4 and C5 sugar rings to produce alginate oligosaccharides consisting of 2-25 monomers, which have various physiological functions. Thus, it can be used for the continuous industrial production of alginate oligosaccharides with a specific degree of polymerization, in accordance with the requirements of green exploitation of marine resources. With the development of structural analysis, the quantity of characterized alginate lyase structures is progressively growing, leading to a concomitant improvement in understanding the catalytic mechanism. Additionally, the use of molecular modification methods including rational design, truncated expression of non-catalytic domains, and recombination of conserved domains can improve the catalytic properties of the original enzyme, enabling researchers to screen out the enzyme with the expected excellent performance with high success rate and less workload. This review presents the latest findings on the catalytic mechanism of alginate lyases and outlines the methods for molecular modifications. Moreover, it explores the connection between the degree of polymerization and the physiological functions of alginate oligosaccharides, providing a reference for enzymatic preparation development and utilization.
Collapse
Affiliation(s)
- Chen Chen
- The State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Xingfei Li
- The State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Cheng Lu
- The State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; School of Bioengineering, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Xing Zhou
- The State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Long Chen
- The State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Chao Qiu
- The State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Zhengyu Jin
- The State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Jie Long
- The State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
24
|
Zhuang H, Xue P, Shao S, Zeng X, Yan S. In situ generation of hybrid alginate hydrogels for enhanced breast tumor ferrotherapy through multiplex magnifying redox imbalances. Int J Biol Macromol 2024; 258:128952. [PMID: 38143049 DOI: 10.1016/j.ijbiomac.2023.128952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/09/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Ferroptosis has drawn great attention to tumor treatments over the past decade. However, how to specifically boost tumoral redox imbalance by simultaneously superimposing iron-mediated reactive oxygen species and undermining antioxidative pathways at the tumor site is still a significant challenge in ferroptosis-based tumor ferrotherapy. In this study, we designed an in situ generable hydrogel that contains paclitaxel/chlorin e6-loaded iron-based metal-organic framework (Fe-MOF) nanoparticles for enhanced breast tumor ferrotherapy by multiplex magnifying redox imbalance. The polysaccharide sodium alginate can crosslink with tumoral calcium ions to generate a hydrogel patch, which promotes the retention of Fe-MOF and therapeutic molecules. The Fe-MOF holds peroxidase/glutathione oxidase mimicking properties, resulting in OH generation via the Fenton reaction and glutathione consumption. Local ultrasound treatment facilitates the release of therapeutics and stimulates the generation of signet oxygen by activating the sonosensitizer chlorin e6. In the meanwhile, the low-dose paclitaxel reduces tumoral pH value by downregulating the glutaminolysis-related gene (SLC7A11) which in turn enhances the catalytic activity of Fe-MOF and inhibits antioxidative pathways, respectively. Both in vivo and in vitro experiments show that our designed hybrid hydrogels can induce significant ferrotherapeutic effects by augmenting the tumoral oxidative stresses.
Collapse
Affiliation(s)
- Huilan Zhuang
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, The Straits Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, Fujian, China
| | - Panpan Xue
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, The Straits Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, Fujian, China
| | - Sijie Shao
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, The Straits Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, Fujian, China
| | - Xuemei Zeng
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, 1 Keji Road, Fuzhou 350117, China.
| | - Shuangqian Yan
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, The Straits Laboratory of Flexible Electronics (SLoFE), Fuzhou 350117, Fujian, China.
| |
Collapse
|
25
|
Inam A, Oncu-Oner T, Deniz I. Algae in Biomedicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1470:147-163. [PMID: 38353867 DOI: 10.1007/5584_2024_795] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Algae, which live in marine or freshwater, are photosynthetic organisms. They vary greatly in size, morphology, and degree of complexity of their body structures. Algae are generally divided into two main groups, microalgae, which are small in size, and macroalgae, which are larger in size. These aquatic organisms have rich and valuable compounds including sterols, polysaccharides, pigments, fatty acids, proteins, enzymes, minerals, and vitamins that could be used in different application fields due to their bioactive functions. In recent years, algae and their components have attracted interest in biomedicine and health applications as their bioactive components could show antioxidant, anticancer, anti-inflammatory, antiviral, antiangiogenic, antidiabetic, antiobesity, immunostimulatory, vaccine adjuvant, and hypolipidemic activities. In this chapter, these activities and bioactive components underlying these properties are reviewed.
Collapse
Affiliation(s)
- Aysegul Inam
- Bioengineering Department, Faculty of Engineering and Natural Sciences, Manisa Celal Bayar University, Yunusemre-Manisa, Turkey
| | - Tulay Oncu-Oner
- Bioengineering Department, Faculty of Engineering and Natural Sciences, Manisa Celal Bayar University, Yunusemre-Manisa, Turkey
| | - Irem Deniz
- Bioengineering Department, Faculty of Engineering and Natural Sciences, Manisa Celal Bayar University, Yunusemre-Manisa, Turkey.
| |
Collapse
|
26
|
Zhou J, Yang T, Chen Z, Chen Y, Li S. A non-thermal modification method to promote the interaction of zein-alginate oligosaccharides composites for better encapsulation and stability-Cold plasma. Int J Biol Macromol 2023; 253:126496. [PMID: 37633568 DOI: 10.1016/j.ijbiomac.2023.126496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/29/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
This current research explored the application of cold plasma (CP) treatment to modify zein-alginate oligosaccharide (zein-AOS) composites in an ethanol-water solution. Anti-solvent method was used to prepare zein-AOS nanoparticles (NPs), and the objective was to investigate the mechanism by which CP promotes interaction between protein and saccharides. Characterization results indicated that CP treatment improved hydrogen bonding and electrostatic interaction between zein and AOS. The CP zein-AOS NPs underwent dispersion and rearrangement, resulting in smaller aggregates with better dispersibility. Among the various induction conditions tested, the zein-AOS85 NPs (induced at 85 W for 2 min) exhibited superior performance as delivery wall materials, with smaller particle size (234.67 nm), larger specific surface area (9.443 m2/g), and higher surface charge (-35.43 mV). In addition, zein-AOS85 showed high stability when used as delivery wall material, providing more binding sites and self-assembly dynamics for nutrients. Curcumin was used as the nutrient model in this study, and CP was found to enhance hydrogen bonding, electrostatic interaction, and hydrophobic interaction between zein, AOS, and nutrients, resulting in increased encapsulation efficiency (EE) from 63.80 % to 85.17 %. The delivery system also exhibited good pH, ionic strength, storage, and dispersion stability.
Collapse
Affiliation(s)
- Junjun Zhou
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Tongliang Yang
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Ziyang Chen
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Ye Chen
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shuhong Li
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
27
|
Krishna Perumal P, Huang CY, Chen CW, Anisha GS, Singhania RR, Dong CD, Patel AK. Advances in oligosaccharides production from brown seaweeds: extraction, characterization, antimetabolic syndrome, and other potential applications. Bioengineered 2023; 14:2252659. [PMID: 37726874 PMCID: PMC10512857 DOI: 10.1080/21655979.2023.2252659] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/27/2023] [Indexed: 09/21/2023] Open
Abstract
Brown seaweeds are a promising source of bioactive substances, particularly oligosaccharides. This group has recently gained considerable attention due to its diverse cell wall composition, structure, and wide-spectrum bioactivities. This review article provides a comprehensive update on advances in oligosaccharides (OSs) production from brown seaweeds and their potential health applications. It focuses on advances in feedstock pretreatment, extraction, characterization, and purification prior to OS use for potential health applications. Brown seaweed oligosaccharides (BSOSs) are extracted using various methods. Among these, enzymatic hydrolysis is the most preferred, with high specificity, mild reaction conditions, and low energy consumption. However, the enzyme selection and hydrolysis conditions need to be optimized for desirable yield and oligosaccharides composition. Characterization of oligosaccharides is essential to determine their structure and properties related to bioactivities and to predict their most suitable application. This is well covered in this review. Analytical techniques such as high-performance liquid chromatography (HPLC), gas chromatography (GC), and nuclear magnetic resonance (NMR) spectroscopy are commonly applied to analyze oligosaccharides. BSOSs exhibit a range of biological properties, mainly antimicrobial, anti-inflammatory, and prebiotic properties among others. Importantly, BSOSs have been linked to possible health advantages, including metabolic syndrome management. Metabolic syndrome is a cluster of conditions, such as obesity, hypertension, and dyslipidemia, which increase the risk of cardiovascular disease and type 2 diabetes. Furthermore, oligosaccharides have potential applications in the food and pharmaceutical industries. Future research should focus on improving industrial-scale oligosaccharide extraction and purification, as well as researching their potential utility in the treatment of various health disorders.[Figure: see text].
Collapse
Affiliation(s)
- Pitchurajan Krishna Perumal
- Institute of Aquatic Science and Technology, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Chun-Yung Huang
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Chiu-Wen Chen
- Institute of Aquatic Science and Technology, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Sustainable Environment Research Center, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Department of Marine Environmental Engineering, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Grace Sathyanesan Anisha
- Post-Graduate and Research Department of Zoology, Government College for Women, Thiruvananthapuram, India
| | - Reeta Rani Singhania
- Institute of Aquatic Science and Technology, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Centre for Energy and Environmental Sustainability, Lucknow, Uttar Pradesh, India
| | - Cheng-Di Dong
- Sustainable Environment Research Center, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Department of Marine Environmental Engineering, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Centre for Energy and Environmental Sustainability, Lucknow, Uttar Pradesh, India
| | - Anil Kumar Patel
- Institute of Aquatic Science and Technology, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Centre for Energy and Environmental Sustainability, Lucknow, Uttar Pradesh, India
| |
Collapse
|
28
|
Zhang E, Shang C, Ma M, Zhang X, Liu Y, Song S, Li X. Polyguluronic acid alleviates doxorubicin-induced cardiotoxicity by suppressing Peli1-NLRP3 inflammasome-mediated pyroptosis. Carbohydr Polym 2023; 321:121334. [PMID: 37739547 DOI: 10.1016/j.carbpol.2023.121334] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/24/2023]
Abstract
Polyguluronic acid (PG), a polysaccharide from alginate, possesses excellent bioactivities. We prepared high-purity PG with 10.41 kDa molecular weight (Mw) and a 59 average degree of polymerization (DP) by acid hydrolysis, three pH grades, Q-Sepharose column elution, and Sephadex G-25 column desalination. Then, we evaluated the PG protective effects on doxorubicin-induced cardiotoxicity (DIC) in vitro and in vivo. The nontoxic PG enhanced cellular viability, reduced cell pyroptosis morphology, diminished the LDH and IL-1β release, and downregulated expressions of ASC oligomerization, NLRP3, cl-CASP1, and GSDMD, by which PG protected the cardiomyocytes from NLRP3 inflammasome-mediated pyroptosis in doxorubicin-stimulated HL-1 cells and C57BL/6J mice. The probable underlying mechanism may be that PG downregulated doxorubicin -induced Peli1, the deficiency of which could inhibit doxorubicin-induced NLRP3 inflammasome-mediated pyroptosis. These results suggested that polysaccharide PG from alginate could prevent DIC and may be a potential therapeutic agent or bioactive material for preventing DIC.
Collapse
Affiliation(s)
- E Zhang
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Chuangeng Shang
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Mingtao Ma
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Xuanfeng Zhang
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Yu Liu
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Shuliang Song
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Xia Li
- Marine College, Shandong University, Weihai, Shandong 264209, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
29
|
Zueva OS, Khair T, Kazantseva MA, Latypova L, Zuev YF. Ions-Induced Alginate Gelation According to Elemental Analysis and a Combinatorial Approach. Int J Mol Sci 2023; 24:16201. [PMID: 38003391 PMCID: PMC10671519 DOI: 10.3390/ijms242216201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
This study considers the potential of elemental analysis of polysaccharide ionotropic gels in elucidating the junction zones for different divalent cations. The developed algorithm ensures the correct separation of contributions from physically adsorbed and structure-forming ionic compounds, with the obtained results scaled to alginate C12 block. Possible versions of chain association into dimers and their subsequent integration into flat junction zones were analyzed within the framework of the "egg-box" model. The application of combinatorial analysis made it possible to derive theoretical relations to find the probability of various types of egg-box cell occurrences for alginate chains with arbitrary monomeric units ratio μ = M/G, which makes it possible to compare experimental data for alginates of different origins. Based on literature data and obtained chemical formulas, the possible correspondence of concrete biopolymer cells to those most preferable for filling by alkaline earth cations was established. The identified features of elemental composition suggest the formation of composite hydrated complexes with the participation of transition metal cations. The possibility of quantitatively assessing ordered secondary structures formed due to the physical sorption of ions and molecules from environment, correlating with the sorption capabilities of Me2+ alginate, was established.
Collapse
Affiliation(s)
- Olga S. Zueva
- Institute of Electric Power Engineering and Electronics, Kazan State Power Engineering University, 51 Krasnoselskaya Street, 420066 Kazan, Russia; (O.S.Z.); (T.K.)
| | - Tahar Khair
- Institute of Electric Power Engineering and Electronics, Kazan State Power Engineering University, 51 Krasnoselskaya Street, 420066 Kazan, Russia; (O.S.Z.); (T.K.)
| | - Mariia A. Kazantseva
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky Street, 420111 Kazan, Russia;
- School of Applied Mathematics, HSE University, 34 Tallinskaya Street, 123458 Moscow, Russia
| | - Larisa Latypova
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 92 West Da-Zhi Street, Harbin 150001, China;
| | - Yuriy F. Zuev
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky Street, 420111 Kazan, Russia;
| |
Collapse
|
30
|
Gruber JV, Holtz R. Living, quiescent Lactobacillus plantarum Lp90 probiotic, delivered topically to full thickness tissues in vitro via a just-add-water cream delivery system, stimulates the expression of elastin protein. J Cosmet Dermatol 2023; 22:2852-2860. [PMID: 37470208 DOI: 10.1111/jocd.15927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/08/2023] [Accepted: 06/29/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Delivering living probiotics to the skin can be challenging as most water-containing cosmetic products require preservatives to maintain product stability. A recently introduced powdered technology [Stratabiosys™, Vantage Personal Care] allows for quiescent probiotic powders to be stored for extended periods of time. The powders can then be reconstituted to creams at the point of use by adding water and mixing and were examined in vitro on reconstructed human full thickness tissues to see if the probiotic had any influence of several important biomolecules expressed in the skin. MATERIALS AND METHODS A probiotic powder containing 200 M CFU/gram of living quiescent Lactobacillus plantarum Lp90 was reconstituted to a cream by adding ultrapure water and gently mixing the components at room temperature to quickly produce a cream. The resulting cream was tested topically on Epiderm® Full Thickness Tissues by treating the tissues for 24 h, removing the cream with a PBS rinse and then repeating the treatment for another 24 h. The resulting tissues were examined for four strategically important skin biomolecules including Type 1A collagen, elastin, filaggrin and hyaluronic acid. The probiotic-containing powder was tested against untreated tissues and powders not containing probiotics and powders containing measured amounts of one of two cryoprotectants known to be used to maintain the integrity of the quiescent probiotics during drying of the quiescent probiotic powders. RESULTS It was found that topical treatment on Epiderm® tissues with creams containing 2 M (1%), 4 M (2%) and 6 M (3%) CFU/gram prepared from a base powder containing 200 M CFU/gram of Lactobacillus plantarum Lp90 stimulated elastin expression in a dose dependent fashion. There was no effect on the other biomolecules examined in the studies. In addition, it was found that creams made from powders containing only the known cryoprotectants, not bacteria, had no influence on elastin expression. CONCLUSION The results of this study demonstrate that topical delivery of probiotics is possible from powders containing quiescent probiotic powders converted to creams just prior to application to the tissues. In the case of a powder containing Lactobacillus plantarum Lp90, topical application significantly increased expression of elastin in the skin replicants after 48 h of exposure to the cream made with the probiotic. The elastin-stimulating effects are not coming from the oligosaccharide cryoprotectants used to maintain the probiotic powders in their quiescent, dried state. The results indicate that it is the living Lactobacillus plantarum probiotic that is stimulating the elastin expression in the skin tissues.
Collapse
Affiliation(s)
| | - Robert Holtz
- Bioinnovation Laboratories, Inc., Lakewood, Colorado, USA
| |
Collapse
|
31
|
Krishna Perumal P, Dong CD, Chauhan AS, Anisha GS, Kadri MS, Chen CW, Singhania RR, Patel AK. Advances in oligosaccharides production from algal sources and potential applications. Biotechnol Adv 2023; 67:108195. [PMID: 37315876 DOI: 10.1016/j.biotechadv.2023.108195] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023]
Abstract
In recent years, algal-derived glycans and oligosaccharides have become increasingly important in health applications due to higher bioactivities than plant-derived oligosaccharides. The marine organisms have complex, and highly branched glycans and more reactive groups to elicit greater bioactivities. However, complex and large molecules have limited use in broad commercial applications due to dissolution limitations. In comparison to these, oligosaccharides show better solubility and retain their bioactivities, hence, offering better applications opportunity. Accordingly, efforts are being made to develop a cost-effective method for enzymatic extraction of oligosaccharides from algal polysaccharides and algal biomass. Yet detailed structural characterization of algal-derived glycans is required to produce and characterize the potential biomolecules for improved bioactivity and commercial applications. Some macroalgae and microalgae are being evaluated as in vivo biofactories for efficient clinical trials, which could be very helpful in understanding the therapeutic responses. This review discusses the recent advancements in the production of oligosaccharides from microalgae. It also discusses the bottlenecks of the oligosaccharides research, technological limitations, and probable solutions to these problems. Furthermore, it presents the emerging bioactivities of algal oligosaccharides and their promising potential for possible biotherapeutic application.
Collapse
Affiliation(s)
- Pitchurajan Krishna Perumal
- Institute of Aquatic Science and Technology, National Kaohsiung University of Science and Technology, Kaohsiung City 81157, Taiwan
| | - Cheng-Di Dong
- Institute of Aquatic Science and Technology, National Kaohsiung University of Science and Technology, Kaohsiung City 81157, Taiwan; Sustainable Environment Research Centre, National Kaohsiung University of Science and Technology, Kaohsiung City 81157, Taiwan; Department of Marine Environmental Engineering, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Ajeet Singh Chauhan
- Institute of Aquatic Science and Technology, National Kaohsiung University of Science and Technology, Kaohsiung City 81157, Taiwan
| | - Grace Sathyanesan Anisha
- Post-Graduate and Research Department of Zoology, Government College for Women, Thiruvananthapuram 695014, Kerala, India
| | - Mohammad Sibtain Kadri
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung City-804201, Taiwan
| | - Chiu-Wen Chen
- Institute of Aquatic Science and Technology, National Kaohsiung University of Science and Technology, Kaohsiung City 81157, Taiwan; Sustainable Environment Research Centre, National Kaohsiung University of Science and Technology, Kaohsiung City 81157, Taiwan; Department of Marine Environmental Engineering, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Reeta Rani Singhania
- Institute of Aquatic Science and Technology, National Kaohsiung University of Science and Technology, Kaohsiung City 81157, Taiwan; Centre for Energy and Environmental Sustainability, Lucknow 226 029, Uttar Pradesh, India
| | - Anil Kumar Patel
- Institute of Aquatic Science and Technology, National Kaohsiung University of Science and Technology, Kaohsiung City 81157, Taiwan; Centre for Energy and Environmental Sustainability, Lucknow 226 029, Uttar Pradesh, India.
| |
Collapse
|
32
|
Guo Q, Dan M, Zheng Y, Shen J, Zhao G, Wang D. Improving the thermostability of a novel PL-6 family alginate lyase by rational design engineering for industrial preparation of alginate oligosaccharides. Int J Biol Macromol 2023; 249:125998. [PMID: 37499708 DOI: 10.1016/j.ijbiomac.2023.125998] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Alginate is degraded into alginate oligosaccharides with various biological activities by enzymes. However, the thermostability of the enzyme limits its industrial application. In this study, a novel PL-6 alginate lyase, AlyRm6A from Rhodothermus marinus 4252 was expressed and characterized. In addition, an efficient comprehensive strategy was proposed, including automatic design of heat-resistant mutants, multiple computer-aided ΔΔGfold value calculation, and conservative analysis of mutation sites. AlyRm6A has naturally high thermostability. Compared with the WT, T43I and Q216I kept their original activities, and their half-lives were increased from 3.68 h to 4.29 h and 4.54 h, melting point temperatures increased from 61.5 °C to 62.9 °C and 63.5 °C, respectively. The results of circular dichroism showed that both the mutants and the wild type had the characteristic peaks of β-sheet at 195 nm and 216 nm, which indicated that there was no significant effect on the secondary structure of the protein. Molecular dynamics simulation (MD) analyses suggest that the enhancement of the hydrophobic interaction network, improvement of molecular rigidity, and denser structure could improve the stability of AlyRm6A. To the best of our knowledge, our findings indicate that AlyRm6A mutants exhibit the highest thermostability among the characterized PL-6 alginate lyases, making them potential candidates for industrial production of alginate oligosaccharides.
Collapse
Affiliation(s)
- Qing Guo
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Meiling Dan
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Yuting Zheng
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Ji Shen
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Guohua Zhao
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Damao Wang
- College of Food Science, Southwest University, Chongqing 400715, China.
| |
Collapse
|
33
|
Du YW, Liu L, Feng NJ, Zheng DF, Liu ML, Zhou H, Deng P, Wang YX, Zhao HM. Combined transcriptomic and metabolomic analysis of alginate oligosaccharides alleviating salt stress in rice seedlings. BMC PLANT BIOLOGY 2023; 23:455. [PMID: 37770835 PMCID: PMC10540332 DOI: 10.1186/s12870-023-04470-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023]
Abstract
BACKGROUND Salt stress is one of the key factors limiting rice production. Alginate oligosaccharides (AOS) enhance plant stress resistance. However, the molecular mechanism underlying salt tolerance in rice induced by AOS remains unclear. FL478, which is a salt-tolerant indica recombinant inbred line and IR29, a salt-sensitive rice cultivar, were used to comprehensively analyze the effects of AOS sprayed on leaves in terms of transcriptomic and metabolite profiles of rice seedlings under salt stress. RESULTS In this experiment, exogenous application of AOS increased SOD, CAT and APX activities, as well as GSH and ASA levels to reduce the damage to leaf membrane, increased rice stem diameter, the number of root tips, aboveground and subterranean biomass, and improved rice salt tolerance. Comparative transcriptomic analyses showed that the regulation of AOS combined with salt treatment induced the differential expression of 305 and 1030 genes in FL478 and IR29. The expressed genes enriched in KEGG pathway analysis were associated with antioxidant levels, photosynthesis, cell wall synthesis, and signal transduction. The genes associated with light-trapping proteins and RLCK receptor cytoplasmic kinases, including CBA, LHCB, and Lhcp genes, were fregulated in response to salt stress. Treatment with AOS combined with salt induced the differential expression of 22 and 50 metabolites in FL478 and IR29. These metabolites were mainly related to the metabolism of amino and nucleotide sugars, tryptophan, histidine, and β -alanine. The abundance of metabolites associated with antioxidant activity, such as 6-hydroxymelatonin, wedelolactone and L-histidine increased significantly. Combined transcriptomic and metabolomic analyses revealed that dehydroascorbic acid in the glutathione and ascorbic acid cycles plays a vital role in salt tolerance mediated by AOS. CONCLUSION AOS activate signal transduction, regulate photosynthesis, cell wall formation, and multiple antioxidant pathways in response to salt stress. This study provides a molecular basis for the alleviation of salt stress-induced damage by AOS in rice.
Collapse
Affiliation(s)
- You-Wei Du
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China
| | - Ling Liu
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China
| | - Nai-Jie Feng
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China.
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China.
- Shenzhen Research Institute of Guangdong Ocean University, Shenzhen, 518108, China.
| | - Dian-Feng Zheng
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China.
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China.
- Shenzhen Research Institute of Guangdong Ocean University, Shenzhen, 518108, China.
| | - Mei-Ling Liu
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China
| | - Hang Zhou
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China
| | - Peng Deng
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China
| | - Ya-Xing Wang
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China
| | - Hui-Min Zhao
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
- South China Center of National Saline-tolerant Rice Technology Innovation Center, Zhanjiang, 524088, China
| |
Collapse
|
34
|
Miao W, Jiang H, Li X, Sang S, Jiang L, Lin Q, Zhang Z, Chen L, Long J, Jiao A, Wang J, Jin Z, Qiu C. Recent advances in natural gums as additives to help the construction and application of edible biopolymer gels: the example of hydrogels and oleogels. Crit Rev Food Sci Nutr 2023; 64:12702-12719. [PMID: 37702742 DOI: 10.1080/10408398.2023.2256006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Novel, innovative approaches like edible gels (hydrogels and oleogels) are important food materials with great scientific interest due to their positive impacts on structural and functional foods and other unique properties. Biopolymers (protein, starch and other polysaccharides) can be excellent and cost-effective materials for the formed edible gels. Recently, natural gums, although also as biopolymers, are preferred as additives to further improve the textural and functional properties of edible gels, which have received extensive attention. However, these studies have not been outlined in previous reviews. In this review, we highlighted the advantages of gums as additives to construct edible gels. Moreover, the various roles (including electrostatic or covalent interactions) for natural gums in regulation of food gel properties (solvent-holding and rheological properties) are highlighted. Finally, the use of natural gums as additives to improve the stability and targeted delivery of phytochemicals in food gels and their application in food systems are summarized. The information covered in this article may be useful for the design of functional foods that can better meet personalized needs of people.
Collapse
Affiliation(s)
- Wenbo Miao
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Han Jiang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaojing Li
- College of Light Industry and Food Engineering, Nanjing Forestry University, Jiangsu, China
| | - Shangyuan Sang
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, School of Medicine, Ningbo University, Ningbo, China
| | - Liming Jiang
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, School of Medicine, Ningbo University, Ningbo, China
| | - Qianzhu Lin
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhiheng Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Long Chen
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Jie Long
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Aiquan Jiao
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Jinpeng Wang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhengyu Jin
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Chao Qiu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Collaborative innovation center of food safety and quality control in Jiangsu Province, International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
35
|
Pritchard MF, Powell LC, Adams JYM, Menzies G, Khan S, Tøndervik A, Sletta H, Aarstad O, Skjåk-Bræk G, McKenna S, Buurma NJ, Farnell DJJ, Rye PD, Hill KE, Thomas DW. Structure-Activity Relationships of Low Molecular Weight Alginate Oligosaccharide Therapy against Pseudomonas aeruginosa. Biomolecules 2023; 13:1366. [PMID: 37759766 PMCID: PMC10527064 DOI: 10.3390/biom13091366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/26/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Low molecular weight alginate oligosaccharides have been shown to exhibit anti-microbial activity against a range of multi-drug resistant bacteria, including Pseudomonas aeruginosa. Previous studies suggested that the disruption of calcium (Ca2+)-DNA binding within bacterial biofilms and dysregulation of quorum sensing (QS) were key factors in these observed effects. To further investigate the contribution of Ca2+ binding, G-block (OligoG) and M-block alginate oligosaccharides (OligoM) with comparable average size DPn 19 but contrasting Ca2+ binding properties were prepared. Fourier-transform infrared spectroscopy demonstrated prolonged binding of alginate oligosaccharides to the pseudomonal cell membrane even after hydrodynamic shear treatment. Molecular dynamics simulations and isothermal titration calorimetry revealed that OligoG exhibited stronger interactions with bacterial LPS than OligoM, although this difference was not mirrored by differential reductions in bacterial growth. While confocal laser scanning microscopy showed that both agents demonstrated similar dose-dependent reductions in biofilm formation, OligoG exhibited a stronger QS inhibitory effect and increased potentiation of the antibiotic azithromycin in minimum inhibitory concentration and biofilm assays. This study demonstrates that the anti-microbial effects of alginate oligosaccharides are not purely influenced by Ca2+-dependent processes but also by electrostatic interactions that are common to both G-block and M-block structures.
Collapse
Affiliation(s)
- Manon F. Pritchard
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Lydia C. Powell
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
- Microbiology and Infectious Disease Group, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Georgina Menzies
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK;
| | - Saira Khan
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Anne Tøndervik
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway; (A.T.); (H.S.)
| | - Håvard Sletta
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway; (A.T.); (H.S.)
| | - Olav Aarstad
- Department of Biotechnology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway; (O.A.); (G.S.-B.)
| | - Gudmund Skjåk-Bræk
- Department of Biotechnology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway; (O.A.); (G.S.-B.)
| | - Stephen McKenna
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Niklaas J. Buurma
- Physical Organic Chemistry Centre, School of Chemistry, Cardiff University, Cardiff CF10 3AT, UK;
| | - Damian J. J. Farnell
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Philip D. Rye
- AlgiPharma AS, Industriveien 33, N-1337 Sandvika, Norway;
| | - Katja E. Hill
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - David W. Thomas
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| |
Collapse
|
36
|
Li L, Zhu B, Yao Z, Jiang J. Directed preparation, structure-activity relationship and applications of alginate oligosaccharides with specific structures: A systematic review. Food Res Int 2023; 170:112990. [PMID: 37316063 DOI: 10.1016/j.foodres.2023.112990] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023]
Abstract
The alginate oligosaccharides (AOS) possess versatile activities (such as antioxidant, anti-inflammatory, antitumor, and immune-regulatory activities) and have been the research topic in marine bioresource utilization fields. The degree of polymerization (DP) and the β-D-mannuronic acid (M)/α-L-guluronic acid (G)-units ratio strongly affect the functionality of AOS. Therefore, directed preparation of AOS with specific structures is essential for expanding the applications of alginate polysaccharides and has been the research topic in the marine bioresource field. Alginate lyases could efficiently degrade alginate and specifically produce AOS with specific structures. Therefore, enzymatic preparation of AOS with specific structures has drawn increasing attention. Herein, we systematically summarized the current research progress on the structure-function relation of AOS and focuses on the application of the enzymatic properties of alginate lyase to the specific preparation of various types of AOS. At the same time, current challenges and opportunities for AOS applications are presented to guide and improve the preparation and application of AOS in the future.
Collapse
Affiliation(s)
- Li Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Benwei Zhu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| | - Zhong Yao
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Jinju Jiang
- State Key Laboratory of Bioactive Seaweed Substances, Qingdao 266400, China
| |
Collapse
|
37
|
Cheong KL, Zhang Y, Li Z, Li T, Ou Y, Shen J, Zhong S, Tan K. Role of Polysaccharides from Marine Seaweed as Feed Additives for Methane Mitigation in Ruminants: A Critical Review. Polymers (Basel) 2023; 15:3153. [PMID: 37571046 PMCID: PMC10420924 DOI: 10.3390/polym15153153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Given the increasing concerns regarding greenhouse gas emissions associated with livestock production, the need to discover effective strategies to mitigate methane production in ruminants is clear. Marine algal polysaccharides have emerged as a promising research avenue because of their abundance and sustainability. Polysaccharides, such as alginate, laminaran, and fucoidan, which are extracted from marine seaweeds, have demonstrated the potential to reduce methane emissions by influencing the microbial populations in the rumen. This comprehensive review extensively examines the available literature and considers the effectiveness, challenges, and prospects of using marine seaweed polysaccharides as feed additives. The findings emphasise that marine algal polysaccharides can modulate rumen fermentation, promote the growth of beneficial microorganisms, and inhibit methanogenic archaea, ultimately leading to decreases in methane emissions. However, we must understand the long-term effects and address the obstacles to practical implementation. Further research is warranted to optimise dosage levels, evaluate potential effects on animal health, and assess economic feasibility. This critical review provides insights for researchers, policymakers, and industry stakeholders dedicated to advancing sustainable livestock production and methane mitigation.
Collapse
Affiliation(s)
- Kit-Leong Cheong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.)
| | - Yiyu Zhang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.)
| | - Zhuoting Li
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.)
| | - Tongtong Li
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.)
| | - Yiqing Ou
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.)
| | - Jiayi Shen
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.)
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.)
| | - Karsoon Tan
- Guangxi Key Laboratory of Beibu Gulf Biodiversity Conservation, Beibu Gulf University, Qinzhou 535000, China
| |
Collapse
|
38
|
Flores-Contreras EA, Araújo RG, Rodríguez-Aguayo AA, Guzmán-Román M, García-Venegas JC, Nájera-Martínez EF, Sosa-Hernández JE, Iqbal HMN, Melchor-Martínez EM, Parra-Saldivar R. Polysaccharides from the Sargassum and Brown Algae Genus: Extraction, Purification, and Their Potential Therapeutic Applications. PLANTS (BASEL, SWITZERLAND) 2023; 12:2445. [PMID: 37447006 DOI: 10.3390/plants12132445] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023]
Abstract
Brown macroalgae represent one of the most proliferative groups of living organisms in aquatic environments. Due to their abundance, they often cause problems in aquatic and terrestrial ecosystems, resulting in health problems in humans and the death of various aquatic species. To resolve this, the application of Sargassum has been sought in different research areas, such as food, pharmaceuticals, and cosmetics, since Sargassum is an easy target for study and simple to obtain. In addition, its high content of biocompounds, such as polysaccharides, phenols, and amino acids, among others, has attracted attention. One of the valuable components of brown macroalgae is their polysaccharides, which present interesting bioactivities, such as antiviral, antimicrobial, and antitumoral, among others. There is a wide variety of methods of extraction currently used to obtain these polysaccharides, such as supercritical fluid extraction (SFE), pressurized liquid extraction (PLE), subcritical water extraction (SCWE), ultrasound-assisted extraction (UAE), enzyme-assisted extraction (EAE), and microwave-assisted extraction (MAE). Therefore, this work covers the most current information on the methods of extraction, as well as the purification used to obtain a polysaccharide from Sargassum that is able to be utilized as alginates, fucoidans, and laminarins. In addition, a compilation of bioactivities involving brown algae polysaccharides in in vivo and in vitro studies is also presented, along with challenges in the research and marketing of Sargassum-based products that are commercially available.
Collapse
Affiliation(s)
- Elda A Flores-Contreras
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | - Rafael G Araújo
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | | | - Muriel Guzmán-Román
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | | | - Erik Francisco Nájera-Martínez
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | - Juan Eduardo Sosa-Hernández
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | - Elda M Melchor-Martínez
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | - Roberto Parra-Saldivar
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| |
Collapse
|
39
|
Li Q, Liu X, Yan C, Zhao B, Zhao Y, Yang L, Shi M, Yu H, Li X, Luo K. Polysaccharide-Based Stimulus-Responsive Nanomedicines for Combination Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206211. [PMID: 36890780 DOI: 10.1002/smll.202206211] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/09/2023] [Indexed: 06/08/2023]
Abstract
Cancer immunotherapy is a promising antitumor approach, whereas nontherapeutic side effects, tumor microenvironment (TME) intricacy, and low tumor immunogenicity limit its therapeutic efficacy. In recent years, combination immunotherapy with other therapies has been proven to considerably increase antitumor efficacy. However, achieving codelivery of the drugs to the tumor site remains a major challenge. Stimulus-responsive nanodelivery systems show controlled drug delivery and precise drug release. Polysaccharides, a family of potential biomaterials, are widely used in the development of stimulus-responsive nanomedicines due to their unique physicochemical properties, biocompatibility, and modifiability. Here, the antitumor activity of polysaccharides and several combined immunotherapy strategies (e.g., immunotherapy combined with chemotherapy, photodynamic therapy, or photothermal therapy) are summarized. More importantly, the recent progress of polysaccharide-based stimulus-responsive nanomedicines for combination cancer immunotherapy is discussed, with the focus on construction of nanomedicine, targeted delivery, drug release, and enhanced antitumor effects. Finally, the limitations and application prospects of this new field are discussed.
Collapse
Affiliation(s)
- Qiuxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Xing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Chunmei Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Bolin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yuxin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Lu Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Mingyi Shi
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hua Yu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao SAR, 999078, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Kaipei Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| |
Collapse
|
40
|
Li L, Jiang J, Yao Z, Zhu B. Recent advances in the production, properties and applications of alginate oligosaccharides - a mini review. World J Microbiol Biotechnol 2023; 39:207. [PMID: 37221433 DOI: 10.1007/s11274-023-03658-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/20/2023] [Indexed: 05/25/2023]
Abstract
Alginate oligosaccharides (AOS) made from the degradation of alginate, to some extent, makes up for the poor solubility and bioavailability of alginate as a macromolecular substance and possess several beneficial biological activities that are absent in alginate. These properties include prebiotic, glycolipid regulatory, immunomodulatory, antimicrobial, antioxidant, anti-tumor, promoting plant growth and other activities. Consequently, AOS has significant potential for use in the agricultural, biomedical, and food industries, and has been the focus of research in the field of marine biological resources. This review comprehensively covers methods (physical, chemical, and enzymatic methods) for the production of AOS from alginate. More importantly, this paper reviews recent advances in the biological activity and potentially industrial and therapeutic applications of AOS, providing a reference for future research and applications of AOS.
Collapse
Affiliation(s)
- Li Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China
| | - Jinju Jiang
- State Key Laboratory of Bioactive Seaweed Substances, Qingdao, 266400, China
| | - Zhong Yao
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China
| | - Benwei Zhu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China.
| |
Collapse
|
41
|
Lupu A, Gradinaru LM, Gradinaru VR, Bercea M. Diversity of Bioinspired Hydrogels: From Structure to Applications. Gels 2023; 9:gels9050376. [PMID: 37232968 DOI: 10.3390/gels9050376] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Hydrogels are three-dimensional networks with a variety of structures and functions that have a remarkable ability to absorb huge amounts of water or biological fluids. They can incorporate active compounds and release them in a controlled manner. Hydrogels can also be designed to be sensitive to external stimuli: temperature, pH, ionic strength, electrical or magnetic stimuli, specific molecules, etc. Alternative methods for the development of various hydrogels have been outlined in the literature over time. Some hydrogels are toxic and therefore are avoided when obtaining biomaterials, pharmaceuticals, or therapeutic products. Nature is a permanent source of inspiration for new structures and new functionalities of more and more competitive materials. Natural compounds present a series of physico-chemical and biological characteristics suitable for biomaterials, such as biocompatibility, antimicrobial properties, biodegradability, and nontoxicity. Thus, they can generate microenvironments comparable to the intracellular or extracellular matrices in the human body. This paper discusses the main advantages of the presence of biomolecules (polysaccharides, proteins, and polypeptides) in hydrogels. Structural aspects induced by natural compounds and their specific properties are emphasized. The most suitable applications will be highlighted, including drug delivery, self-healing materials for regenerative medicine, cell culture, wound dressings, 3D bioprinting, foods, etc.
Collapse
Affiliation(s)
- Alexandra Lupu
- "Petru Poni" Institute of Macromolecular Chemistry, 41-A Grigore Ghica Voda Alley, 700487 Iasi, Romania
| | - Luiza Madalina Gradinaru
- "Petru Poni" Institute of Macromolecular Chemistry, 41-A Grigore Ghica Voda Alley, 700487 Iasi, Romania
| | - Vasile Robert Gradinaru
- Faculty of Chemistry, "Alexandru Ioan Cuza" University, 11 Carol I Bd., 700506 Iasi, Romania
| | - Maria Bercea
- "Petru Poni" Institute of Macromolecular Chemistry, 41-A Grigore Ghica Voda Alley, 700487 Iasi, Romania
| |
Collapse
|
42
|
Zhang S, Dong J, Pan R, Xu Z, Li M, Zang R. Structures, Properties, and Bioengineering Applications of Alginates and Hyaluronic Acid. Polymers (Basel) 2023; 15:2149. [PMID: 37177293 PMCID: PMC10181120 DOI: 10.3390/polym15092149] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
In recent years, polymeric materials have been used in a wide range of applications in a variety of fields. In particular, in the field of bioengineering, the use of natural biomaterials offers a possible new avenue for the development of products with better biocompatibility, biodegradability, and non-toxicity. This paper reviews the structural and physicochemical properties of alginate and hyaluronic acid, as well as the applications of the modified cross-linked derivatives in tissue engineering and drug delivery. This paper summarizes the application of alginate and hyaluronic acid in bone tissue engineering, wound dressings, and drug carriers. We provide some ideas on how to replace or combine alginate-based composites with hyaluronic-acid-based composites in tissue engineering and drug delivery to achieve better eco-economic value.
Collapse
Affiliation(s)
- Shuping Zhang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.D.)
| | | | | | | | | | | |
Collapse
|
43
|
Zhang J, Ye X, Li W, Lin Z, Wang W, Chen L, Li Q, Xie X, Xu X, Lu Y. Copper-containing chitosan-based hydrogels enabled 3D-printed scaffolds to accelerate bone repair and eliminate MRSA-related infection. Int J Biol Macromol 2023; 240:124463. [PMID: 37076063 DOI: 10.1016/j.ijbiomac.2023.124463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Bone defect combined with drug-resistant bacteria-related infection is a thorny challenge in clinic. Herein, 3D-printed polyhydroxyalkanoates/β-tricalcium phosphate (PHA/β-TCP, PT) scaffolds were prepared by fused deposition modeling. Then copper-containing carboxymethyl chitosan/alginate (CA/Cu) hydrogels were integrated with the scaffolds via a facile and low-cost chemical crosslinking method. The resultant PT/CA/Cu scaffolds could not only promote proliferation but also osteogenic differentiation of preosteoblasts in vitro. Moreover, PT/CA/Cu scaffolds exhibited a strong antibacterial activity towards a broad-spectrum of bacteria including methicillin-resistant Staphylococcus aureus (MRSA) through inducing the intercellular generation of reactive oxygen species. In vivo experiments further demonstrated that PT/CA/Cu scaffolds significantly accelerated bone repair of cranial defects and efficiently eliminated MRSA-related infection, showing potential for application in infected bone defect therapy.
Collapse
Affiliation(s)
- Jinwei Zhang
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Key Lab of Orthopedic Technology and Implant Materials, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - Xiangling Ye
- Department of Orthopedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330006, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Department of Orthopedics, Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou, Guangdong 510095, China
| | - Wenhua Li
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zefeng Lin
- Guangdong Key Lab of Orthopedic Technology and Implant Materials, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - Wanshun Wang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Lingling Chen
- Guangdong Key Lab of Orthopedic Technology and Implant Materials, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - Qi Li
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaobo Xie
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Xuemeng Xu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China; Department of Orthopedics, Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou, Guangdong 510095, China.
| | - Yao Lu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Key Lab of Orthopedic Technology and Implant Materials, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China.
| |
Collapse
|
44
|
Jiang G, Wang S, Xie J, Tan P, Han L. Discontinuous low temperature stress and plant growth regulators during the germination period promote roots growth in alfalfa (Medicago sativa L.). PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2023; 197:107624. [PMID: 36948023 DOI: 10.1016/j.plaphy.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/15/2023] [Accepted: 03/01/2023] [Indexed: 06/18/2023]
Abstract
In high-cold regions, alfalfa is susceptible to cold damage during the seed germination. The effects of discontinuous low temperature stress and plant growth regulators (PGRs) on alfalfa were studied in response to the high day/night temperature differentials in the area. The experiments included seed germination, seedling cold tolerance and plant recovery. Variable temperatures (VT) of 0 °C/15 °C, 5 °C/20 °C and 10 °C/25 °C were set and seeds were soaked with alginate oligosaccharides (AOS), brassinolide (BR) and diethyl aminoethyl hexanoate (DA-6) during the germination period. Parameters such as seed germination and mean germination time (MGT), phenylalanine ammonia-lyase (PAL) activity and oligomeric proanthocyanidins (OPC) content of early seedlings, dry matter accumulation and root crown of the restored plants were analysed. The results showed that low variable-temperature (LVT) stress prolonged the MGT but had little inhibitory effect on germination percentage. Early seedlings adapted to LVT stress by regulating their own water and OPC content, PAL activity and other parameters. LVT induced early alfalfa seedlings to increase their underground biomass by shortening root length and increasing root diameter, and those that had accumulated more underground biomass had faster growth rates and higher total biomass when the ambient temperature rose. AOS also promoted an increase in root crown diameter and root dry weight. This research proved that LVT stress and AOS during the germination process can lead to better growth of alfalfa in high cold regions.
Collapse
Affiliation(s)
- Gaoqian Jiang
- Institute of Genetics and Developmental Biology Center for Agricultural Resources Research, Chinese Academy of Sciences / Hebei Key Laboratory of Soil Ecology / Key Laboratory of Agricultural Water Resources, Chinese Academy of Sciences, Shijiazhuang, 050022, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shichao Wang
- Institute of Genetics and Developmental Biology Center for Agricultural Resources Research, Chinese Academy of Sciences / Hebei Key Laboratory of Soil Ecology / Key Laboratory of Agricultural Water Resources, Chinese Academy of Sciences, Shijiazhuang, 050022, China
| | - Jin Xie
- Institute of Genetics and Developmental Biology Center for Agricultural Resources Research, Chinese Academy of Sciences / Hebei Key Laboratory of Soil Ecology / Key Laboratory of Agricultural Water Resources, Chinese Academy of Sciences, Shijiazhuang, 050022, China
| | - Pan Tan
- Institute of Genetics and Developmental Biology Center for Agricultural Resources Research, Chinese Academy of Sciences / Hebei Key Laboratory of Soil Ecology / Key Laboratory of Agricultural Water Resources, Chinese Academy of Sciences, Shijiazhuang, 050022, China; University of Chinese Academy of Sciences, Beijing, China
| | - Lipu Han
- Institute of Genetics and Developmental Biology Center for Agricultural Resources Research, Chinese Academy of Sciences / Hebei Key Laboratory of Soil Ecology / Key Laboratory of Agricultural Water Resources, Chinese Academy of Sciences, Shijiazhuang, 050022, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
45
|
Zhou J, Qaing S, Yang B, Wang Y, Wang J, Yang T, Zhang Y, Chen Y, Li S. Cold plasma treatment with alginate oligosaccharide improves the digestive stability and bioavailability of nutrient-delivered particles: An in vitro INFOGEST gastrointestinal study. Int J Biol Macromol 2023; 232:123309. [PMID: 36652987 DOI: 10.1016/j.ijbiomac.2023.123309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023]
Abstract
To improve the stability and bioavailability of the delivered hydrophobic nutrients, the zein-based delivery system was modified by alginate oligosaccharide (AOS), cold plasma (CP) treatments, and synergistically. The digestive behavior of each was investigated in an INFOGEST static in vitro digestion model. The results showed that AOS and CP treatments and their synergistic effects improved the dispersion and stability of the delivery system, leading to a more concentrated particle size distribution and higher particle surface charge. Both CP treatments and AOS increased the release rate of Curcumin (Cur) at small intestine (11.8 % to 20.5 % and 11.8 % to 24.64 %, respectively), and the synergistic effect was higher (11.8 % to 43.84 %). The wall material modified showed a higher encapsulation efficiency of Cur (52.83 % to 85.17 %). Cur release rate measurements showed that the wall material modified could have a positive effect on the slow release of Cur. SDS-page electrophoresis revealed that the slow release was due to the enhanced resistance of wall material to digestive fluids. Thus, treatment with AOS and CP treatments, and the synergism are suitable for modifying zein-based delivery systems for the encapsulation, stabilization, and slow release of hydrophobic nutrients during digestion in the field of functional foods.
Collapse
Affiliation(s)
- Junjun Zhou
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Siqi Qaing
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Bowen Yang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yuhe Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jiake Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Tongliang Yang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yifu Zhang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Ye Chen
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shuhong Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
46
|
Sukhavattanakul P, Pisitsak P, Ummartyotin S, Narain R. Polysaccharides for Medical Technology: Properties and Applications. Macromol Biosci 2023; 23:e2200372. [PMID: 36353915 DOI: 10.1002/mabi.202200372] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/18/2022] [Indexed: 11/12/2022]
Abstract
Over the past decade, the use of polysaccharides has gained tremendous attention in the field of medical technology. They have been applied in various sectors such as tissue engineering, drug delivery system, face mask, and bio-sensing. This review article provides an overview and background of polysaccharides for biomedical uses. Different types of polysaccharides, for example, cellulose and its derivatives, chitin and chitosan, hyaluronic acid, alginate, and pectin are presented. They are fabricated in various forms such as hydrogels, nanoparticles, membranes, and as porous mediums. Successful development and improvement of polysaccharide-based materials will effectively help users to enhance their quality of personal health, decrease cost, and eventually increase the quality of life with respect to sustainability.
Collapse
Affiliation(s)
- Pongpat Sukhavattanakul
- Department of Materials and Textile Technology, Faculty of Science and Technology, Thammasat University, Pathum, Thani, 12120, Thailand
| | - Penwisa Pisitsak
- Department of Materials and Textile Technology, Faculty of Science and Technology, Thammasat University, Pathum, Thani, 12120, Thailand
| | - Sarute Ummartyotin
- Department of Materials and Textile Technology, Faculty of Science and Technology, Thammasat University, Pathum, Thani, 12120, Thailand
| | - Ravin Narain
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, T6G1H9, Canada
| |
Collapse
|
47
|
Zhang Z, Wang X, Li F. An exploration of alginate oligosaccharides modulating intestinal inflammatory networks via gut microbiota. Front Microbiol 2023; 14:1072151. [PMID: 36778853 PMCID: PMC9909292 DOI: 10.3389/fmicb.2023.1072151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Alginate oligosaccharides (AOS) can be obtained by acidolysis and enzymatic hydrolysis. The products obtained by different methods have different structures and physiological functions. AOS have received increasing interest because of their many health-promoting properties. AOS have been reported to exert protective roles for intestinal homeostasis by modulating gut microbiota, which is closely associated with intestinal inflammation, gut barrier strength, bacterial infection, tissue injury, and biological activities. However, the roles of AOS in intestinal inflammation network remain not well understood. A review of published reports may help us to establish the linkage that AOS may improve intestinal inflammation network by affecting T helper type 1 (Th1) Th2, Th9, Th17, Th22 and regulatory T (Treg) cells, and their secreted cytokines [the hub genes of protein-protein interaction networks include interleukin-1 beta (IL-1β), IL-2, IL-4, IL-6, IL-10 and tumor necrosis factor alpha (TNF-α)] via the regulation of probiotics. The potential functional roles of molecular mechanisms are explored in this study. However, the exact mechanism for the direct interaction between AOS and probiotics or pathogenic bacteria is not yet fully understood. AOS receptors may be located on the plasma membrane of gut microbiota and will be a key solution to address such an important issue. The present paper provides a better understanding of the protecting functions of AOS on intestinal inflammation and immunity.
Collapse
Affiliation(s)
- Zhikai Zhang
- Wuzhoufeng Agricultural Science and Technology Co., Ltd., Yantai, China
| | | | | |
Collapse
|
48
|
Wei B, Ren P, Yang R, Gao Y, Tang Q, Xue C, Wang Y. Ameliorative Effect of Mannuronate Oligosaccharides on Hyperuricemic Mice via Promoting Uric Acid Excretion and Modulating Gut Microbiota. Nutrients 2023; 15:nu15020417. [PMID: 36678288 PMCID: PMC9865265 DOI: 10.3390/nu15020417] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Mannuronate oligosaccharide (MOS) is α-D-mannuronic acid polymer with 1,4-glycosidic linkages that possesses beneficial biological properties. The aim of this study was to investigate the hypouricemic effect of MOS in hyperuricemic mice and demonstrate the possible protective mechanisms involved. In this research, 200 mg/kg/day of MOS was orally administered to hyperuricemic mice for four weeks. The results showed that the MOS treatment significantly reduced the serum uric acid (SUA) level from 176.4 ± 7.9 μmol/L to 135.7 ± 10.9 μmol/L (p < 0.05). MOS alleviated the inflammatory response in the kidney. Moreover, MOS promoted uric acid excretion by regulating the protein levels of renal GLUT9, URAT1 and intestinal GLUT9, ABCG2. MOS modulated the gut microbiota in hyperuricemic mice and decreased the levels of Tyzzerella. In addition, research using antibiotic-induced pseudo-sterile mice demonstrated that the gut microbiota played a crucial role in reducing elevated serum uric acid of MOS in mice. In conclusion, MOS may be a potential candidate for alleviating HUA symptoms and regulating gut microbiota.
Collapse
Affiliation(s)
- Biqian Wei
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Pengfei Ren
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Ruzhen Yang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Yuan Gao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
| | - Qingjuan Tang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
- Correspondence: ; Tel.: +86-186-6140-2667
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Yuming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| |
Collapse
|
49
|
Relevance of drying treatment on the extraction of high valuable compounds from invasive brown seaweed Rugulopteryx okamurae. ALGAL RES 2023. [DOI: 10.1016/j.algal.2022.102917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
50
|
Zhang H, Zhou Y, Xu C, Qin X, Guo Z, Wei H, Yu CY. Mediation of synergistic chemotherapy and gene therapy via nanoparticles based on chitosan and ionic polysaccharides. Int J Biol Macromol 2022; 223:290-306. [PMID: 36347370 DOI: 10.1016/j.ijbiomac.2022.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
Nanoparticles (NPs)-based on various ionic polysaccharides, including chitosan, hyaluronic acid, and alginate have been frequently summarized for controlled release applications, however, most of the published reviews, to our knowledge, focused on the delivery of a single therapeutic agent. A comprehensive summarization of the co-delivery of multiple therapeutic agents by the ionic polysaccharides-based NPs, especially on the optimization of the polysaccharide structure for overcoming various extracellular and intracellular barriers toward maximized synergistic effects, to our knowledge, has been rarely explored so far. For this purpose, the strategies used for overcoming various extracellular and intracellular barriers in vivo were introduced first to provide guidance for the rational design of ionic polysaccharides-based NPs with desired features, including long-term circulation, enhanced cellular internalization, controllable drug/gene release, endosomal escape and improved nucleus localization. Next, four preparation strategies were summarized including three physical methods of polyelectrolyte complexation, ionic crosslinking, and self-assembly and a chemical conjugation approach. The challenges and future trends of this rapidly developing field were finally discussed in the concluding remarks. The important guidelines on the rational design of ionic polysaccharides-based NPs for maximized synergistic efficiency drawn in this review will promote the future generation and clinical translation of polysaccharides-based NPs for cancer therapy.
Collapse
Affiliation(s)
- Haitao Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yangchun Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Chenghui Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xuping Qin
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Zifen Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|