1
|
Ren L, Ye H, Fang J, Cao Q, Zhang C, Dong Z, Feng D, Zuo J, Wang W. Xylooligosaccharide interferes with the cell cycle and reduces the antibiotic tolerance of avian pathogenic Escherichia coli by associating with its potential antimetabolic actions. Poult Sci 2024; 103:104405. [PMID: 39426220 PMCID: PMC11536015 DOI: 10.1016/j.psj.2024.104405] [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: 08/14/2024] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024] Open
Abstract
This study aimed to probe if xylooligosaccharide (XOS) could act as an antimetabolite to impact the cell cycle and antibiotic tolerance of avian pathogenic Escherichia coli (APEC). We firstly measured the bacteriostasis of XOS against APEC O78 and its effect on the growth of APEC O78 growing on different medium. Afterwards, the effects of XOS on xylose operon activation along with the cell cycle and antibiotic tolerance of APEC O78 were analyzed. The results showed that XOS caused no inhibitory circle against APEC O78 and did not affect (P > 0.05) the growth of APEC O78 growing on LB medium. Besides, APEC O78 was unable to grow on M9 medium (carbon-free) added with XOS. However, XOS exerted a similar role as xylose in increasing (P < 0.05) the expression of certain xylose operon genes including xylose isomerase (XylA)-encoding gene (xylA) and xylose-binding periplasmic protein (XylF)-encoding gene (xylF) in APEC O78. The molecular docking simulation revealed that the major monomer components (xylobiose, xylotriose and xylotetraose) of XOS had stable binding potentials to both XylA and XylF proteins of E. coli, as supported by the low binding free energy and the formation of considerable hydrogen bonds between them. The subsequent analysis showed that XOS altered certain cell cycle-related genes expression, especially elevated (P < 0.05) nrdB expression and decreased ihfB expression to a degree. Moreover, XOS played a similar role as 2-deoxy-glucose (a glucose analogue serving as a typical antimetabolite) in lowering (P < 0.05) the number of ampicillin-tolerant APEC O78. Collectively, XOS had no direct bacteriostasis against APEC and could not be metabolized/utilized by APEC O78. However, it might become an analogue of xylose and then activate xylose transport- and metabolism-related proteins in APEC O78, thus functioning as a potential antimetabolite and exerting antimetabolic actions. This could at least partially interpret the observed roles of XOS in interfering with the cell cycle and diminishing the antibiotic tolerance of APEC O78. The above findings expand the knowledges about the functions of XOS and provide a basis for exploring novel strategies to reduce the antibiotic tolerance of APEC.
Collapse
Affiliation(s)
- Lulu Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hui Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiarong Fang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qingyun Cao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Changming Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zemin Dong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Dingyuan Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jianjun Zuo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Weiwei Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
2
|
Dubey I, K N, G V, Rohilla G, Lalruatmawii, Naxine P, P J, Rachamalla M, Kushwaha S. Exploring the hypothetical links between environmental pollutants, diet, and the gut-testis axis: The potential role of microbes in male reproductive health. Reprod Toxicol 2024; 130:108732. [PMID: 39395506 DOI: 10.1016/j.reprotox.2024.108732] [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: 07/15/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024]
Abstract
The gut system, commonly referred to as one of the principal organs of the human "superorganism," is a home to trillions of bacteria and serves an essential physiological function in male reproductive failures or infertility. The interaction of the endocrine-immune system and the microbiome facilitates reproduction as a multi-network system. Some recent studies that link gut microbiota to male infertility are questionable. Is the gut-testis axis (GTA) real, and does it affect male infertility? As a result, this review emphasizes the interconnected links between gut health and male reproductive function via changes in gut microbiota. However, a variety of harmful (endocrine disruptors, heavy metals, pollutants, and antibiotics) and favorable (a healthy diet, supplements, and phytoconstituents) elements promote microbiota by causing dysbiosis and symbiosis, respectively, which eventually modify the activities of male reproductive organs and their hormones. The findings of preclinical and clinical studies on the direct and indirect effects of microbiota changes on testicular functions have revealed a viable strategy for exploring the GTA-axis. Although the GTA axis is poorly understood, it may have potential ties to reproductive issues that can be used for therapeutic purposes in the future.
Collapse
Affiliation(s)
- Itishree Dubey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Nandheeswari K
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Vigneshwaran G
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Gourav Rohilla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Lalruatmawii
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Pratik Naxine
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Jayapradha P
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon SK S7N 5E2, Canada
| | - Sapana Kushwaha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India.
| |
Collapse
|
3
|
Naik GARR, Roy AA, Mutalik S, Dhas N. Unleashing the power of polymeric nanoparticles - Creative triumph against antibiotic resistance: A review. Int J Biol Macromol 2024; 278:134977. [PMID: 39187099 DOI: 10.1016/j.ijbiomac.2024.134977] [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: 02/23/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
Antibiotic resistance (ABR) poses a universal concern owing to the widespread use of antibiotics in various sectors. Nanotechnology emerges as a promising solution to combat ABR, offering targeted drug delivery, enhanced bioavailability, reduced toxicity, and stability. This comprehensive review explores concepts of antibiotic resistance, its mechanisms, and multifaceted approaches to combat ABR. The review provides an in-depth exploration of polymeric nanoparticles as advanced drug delivery systems, focusing on strategies for targeting microbial infections and contributing to the fight against ABR. Nanoparticles revolutionize antimicrobial approaches, emphasizing passive and active targeting. The role of various molecules, including small molecules, antimicrobial peptides, proteins, carbohydrates, and stimuli-responsive systems, is being explored in recent research works. The complex comprehension mechanisms of ABR and strategic use of nanotechnology present a promising avenue for advancing antimicrobial tactics, ensuring treatment efficacy, minimizing toxic effects, and mitigating development of ABR. Polymeric nanoparticles, derived from natural or synthetic polymers, are crucial in overcoming ABR. Natural polymers like chitosan and alginate exhibit inherent antibacterial properties, while synthetic polymers such as polylactic acid (PLA), polyethylene glycol (PEG), and polycaprolactone (PCL) can be engineered for specific antibacterial effects. This comprehensive study provides a valuable source of information for researchers, healthcare professionals, and policymakers engaged in the urgent quest to overcome ABR.
Collapse
Affiliation(s)
- Gaurisha Alias Resha Ramnath Naik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Udupi, Karnataka State 576104, India
| | - Amrita Arup Roy
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Udupi, Karnataka State 576104, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Udupi, Karnataka State 576104, India
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Udupi, Karnataka State 576104, India.
| |
Collapse
|
4
|
Petchimuthu R, Venkatesh S, Kannan S, Balakrishnan V. Solid-state fermentation of brown seaweeds for the production of alginate lyase using marine bacterium Enterobacter tabaci RAU2C. Folia Microbiol (Praha) 2024; 69:1083-1093. [PMID: 38401040 DOI: 10.1007/s12223-024-01150-7] [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: 09/06/2023] [Accepted: 02/08/2024] [Indexed: 02/26/2024]
Abstract
Alginate lyases have countless potential for application in industries and medicine particularly as an appealing biocatalyst for the production of biofuels and bioactive oligosaccharides. Solid-state fermentation (SSF) allows improved production of enzymes and consumes less energy compared to submerged fermentation. Seaweeds can serve as the most promising biomass for the production of biochemicals. Alginate present in the seaweed can be used by alginate lyase-producing bacteria to support growth and can secrete alginate lyase. In this perspective, the current study was directed on the bioprocessing of brown seaweeds for the production of alginate lyase using marine bacterial isolate. A novel alginate-degrading marine bacterium Enterobacter tabaci RAU2C which was previously isolated in the laboratory was used for the production of alginate lyase using Sargassum swartzii as a low-cost solid substrate. Process parameters such as inoculum incubation period and moisture content were optimized for alginate lyase production. SSF resulted in 33.56 U/mL of alginate lyase under the static condition maintained with 75% moisture after 4 days. Further, the effect of different buffers, pH, and temperature on alginate lyase activity was also analyzed. An increase in alginate lyase activity was observed with an increase in moisture content from 60 to 75%. Maximum enzyme activity was perceived with phosphate buffer at pH 7 and 37 °C. Further, the residual biomass after SSF could be employed as biofertilizer for plant growth promotion based on the preliminary analysis. To our knowledge, this is the first report stating the usage of seaweed biomass as a substrate for the production of alginate lyase using solid-state fermentation.
Collapse
Affiliation(s)
- Ramya Petchimuthu
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil-626126, Tamilnadu, India
| | - Subharaga Venkatesh
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil-626126, Tamilnadu, India
| | - Suriyalakshmi Kannan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil-626126, Tamilnadu, India
| | - Vanavil Balakrishnan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil-626126, Tamilnadu, India.
| |
Collapse
|
5
|
Bao K, Yang M, Sun Q, Zhang K, Huang H. Genome Analysis of a Potential Novel Vibrio Species Secreting pH- and Thermo-Stable Alginate Lyase and Its Application in Producing Alginate Oligosaccharides. Mar Drugs 2024; 22:414. [PMID: 39330296 PMCID: PMC11433491 DOI: 10.3390/md22090414] [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: 08/05/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Alginate lyase is an attractive biocatalyst that can specifically degrade alginate to produce oligosaccharides, showing great potential for industrial and medicinal applications. Herein, an alginate-degrading strain HB236076 was isolated from Sargassum sp. in Qionghai, Hainan, China. The low 16S rRNA gene sequence identity (<98.4%), ANI value (<71.9%), and dDDH value (<23.9%) clearly indicated that the isolate represented a potential novel species of the genus Vibrio. The genome contained two chromosomes with lengths of 3,007,948 bp and 874,895 bp, respectively, totaling 3,882,843 bp with a G+C content of 46.5%. Among 3482 genes, 3332 protein-coding genes, 116 tRNA, and 34 rRNA sequences were predicted. Analysis of the amino acid sequences showed that the strain encoded 73 carbohydrate-active enzymes (CAZymes), predicting seven PL7 (Alg1-7) and two PL17 family (Alg8, 9) alginate lyases. The extracellular alginate lyase from strain HB236076 showed the maximum activity at 50 °C and pH 7.0, with over 90% activity measured in the range of 30-60 °C and pH 6.0-10.0, exhibiting a wide range of temperature and pH activities. The enzyme also remained at more than 90% of the original activity at a wide pH range (3.0-9.0) and temperature below 50 °C for more than 2 h, demonstrating significant thermal and pH stabilities. Fe2+ had a good promoting effect on the alginate lyase activity at 10 mM, increasing by 3.5 times. Thin layer chromatography (TLC) and electrospray ionization mass spectrometry (ESI-MS) analyses suggested that alginate lyase in fermentation broth could catalyze sodium alginate to produce disaccharides and trisaccharides, which showed antimicrobial activity against Shigella dysenteriae, Aeromonas hydrophila, Staphylococcus aureus, Streptococcus agalactiae, and Escherichia coli. This research provided extended insights into the production mechanism of alginate lyase from Vibrio sp. HB236076, which was beneficial for further application in the preparation of pH-stable and thermo-stable alginate lyase and alginate oligosaccharides.
Collapse
Affiliation(s)
- Ke Bao
- Institute of Tropical Bioscience and Biotechnology, Hainan Institute for Tropical Agricultural Resources, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (K.B.); (M.Y.)
- Hangzhou Watson Biotechnology Co., Ltd., Hangzhou 311400, China;
| | - Miao Yang
- Institute of Tropical Bioscience and Biotechnology, Hainan Institute for Tropical Agricultural Resources, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (K.B.); (M.Y.)
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Qianhuan Sun
- Hangzhou Watson Biotechnology Co., Ltd., Hangzhou 311400, China;
| | - Kaishan Zhang
- Hangzhou Watson Biotechnology Co., Ltd., Hangzhou 311400, China;
| | - Huiqin Huang
- Institute of Tropical Bioscience and Biotechnology, Hainan Institute for Tropical Agricultural Resources, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (K.B.); (M.Y.)
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Rouillard KR, Esther CP, Kissner WJ, Plott LM, Bowman DW, Markovetz MR, Hill DB. Combination treatment to improve mucociliary transport of Pseudomonas aeruginosa biofilms. PLoS One 2024; 19:e0294120. [PMID: 38394229 PMCID: PMC10890754 DOI: 10.1371/journal.pone.0294120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/25/2023] [Indexed: 02/25/2024] Open
Abstract
People with muco-obstructive pulmonary diseases such as cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) often have acute or chronic respiratory infections that are difficult to treat due in part to the accumulation of hyperconcentrated mucus within the airway. Mucus accumulation and obstruction promote chronic inflammation and infection and reduce therapeutic efficacy. Bacterial aggregates in the form of biofilms exhibit increased resistance to mechanical stressors from the immune response (e.g., phagocytosis) and chemical treatments including antibiotics. Herein, combination treatments designed to disrupt the mechanical properties of biofilms and potentiate antibiotic efficacy are investigated against mucus-grown Pseudomonas aeruginosa biofilms and optimized to 1) alter biofilm viscoelastic properties, 2) increase mucociliary transport rates, and 3) reduce bacterial viability. A disulfide bond reducing agent (tris(2-carboxyethyl)phosphine, TCEP), a surfactant (NP40), a biopolymer (hyaluronic acid, HA), a DNA degradation enzyme (DNase), and an antibiotic (tobramycin) are tested in various combinations to maximize biofilm disruption. The viscoelastic properties of biofilms are quantified with particle tracking microrheology and transport rates are quantified in a mucociliary transport device comprised of fully differentiated primary human bronchial epithelial cells. The combination of the NP40 with hyaluronic acid and tobramycin was the most effective at increasing mucociliary transport rates, decreasing the viscoelastic properties of mucus, and reducing bacterial viability. Multimechanistic targeting of biofilm infections may ultimately result in improved clinical outcomes, and the results of this study may be translated into future in vivo infection models.
Collapse
Affiliation(s)
- Kaitlyn R. Rouillard
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | | | - William J. Kissner
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | - Lucas M. Plott
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | - Dean W. Bowman
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | - Matthew R. Markovetz
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | - David B. Hill
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
- Joint Department of Biomedical Engineering, UNC Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
9
|
Li J, Xue C, Shen J, Liu G, Mei X, Sun M, Chang Y. Action Pattern of a Novel G-Specific Alginate Lyase: Determination of Subsite Specificity by HPAEC-PAD/MS. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:1170-1177. [PMID: 38111122 DOI: 10.1021/acs.jafc.3c06778] [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: 12/20/2023]
Abstract
G-specific alginate lyases are important tools for alginate fragment biodegradation and oligosaccharide production, which have great potential in alginate refining research. In this research, a novel G-specific alginate lyase Aly7Ce was cloned, expressed, and characterized, with the optimal reaction conditions at 30 °C and pH 8.0. By employing the UPSEC-VWD-MS method, Aly7Ce was confirmed as a random endoacting alginate lyase. Its minimum substrate was tetrasaccharide, and the final product majorly consisted of disaccharide to tetrasaccharide. HPAEC-PAD/MS method was employed to investigate the structurally different unsaturated alginate oligosaccharides. The substrate recognition and subsite specificity of Aly7Ce were revealed by detecting the oligosaccharide pattern in the enzymatic products with oligosaccharides or polysaccharides as substrates. Aly7Ce mainly attacked the second glycosidic linkage from the nonreducing end of oligosaccharide substrates. The subsite specificity of Aly7Ce was revealed as -2 (M/G), - 1 (G), + 1 (M/G), and +2 (M/G). The regular oligosaccharide products of Aly7Ce could be applied for the efficient preparation of ΔG, ΔGG, and ΔGGG with high purity. The G-specific alginate lyase Aly7Ce with a well-defined product composition and action pattern provided a novel tool for the modification and structural elucidation of alginate, as well as for the targeted preparation of oligosaccharides.
Collapse
Affiliation(s)
- Jiajing Li
- College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao 266404, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao 266404, China
| | - Jingjing Shen
- College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao 266404, China
| | - Guanchen Liu
- College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao 266404, China
| | - Xuanwei Mei
- College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao 266404, China
| | - Menghui Sun
- College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao 266404, China
| | - Yaoguang Chang
- College of Food Science and Engineering, Ocean University of China, 1299 Sansha Road, Qingdao 266404, China
| |
Collapse
|
10
|
Mavrogeni ME, Asadpoor M, Judernatz JH, van Ark I, Wösten MMSM, Strijbis K, Pieters RJ, Folkerts G, Braber S. Protective Effects of Alginate and Chitosan Oligosaccharides against Clostridioides difficile Bacteria and Toxin. Toxins (Basel) 2023; 15:586. [PMID: 37888617 PMCID: PMC10610568 DOI: 10.3390/toxins15100586] [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: 12/27/2022] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 10/28/2023] Open
Abstract
Clostridioides difficile infection is expected to become the most common healthcare-associated infection worldwide. C. difficile-induced pathogenicity is significantly attributed to its enterotoxin, TcdA, which primarily targets Rho-GTPases involved in regulating cytoskeletal and tight junction (TJ) dynamics, thus leading to cytoskeleton breakdown and ultimately increased intestinal permeability. This study investigated whether two non-digestible oligosaccharides (NDOs), alginate (AOS) and chitosan (COS) oligosaccharides, possess antipathogenic and barrier-protective properties against C. difficile bacteria and TcdA toxin, respectively. Both NDOs significantly reduced C. difficile growth, while cell cytotoxicity assays demonstrated that neither COS nor AOS significantly attenuated the TcdA-induced cell death 24 h post-exposure. The challenge of Caco-2 monolayers with increasing TcdA concentrations increased paracellular permeability, as measured by TEER and LY flux assays. In this experimental setup, COS completely abolished, and AOS mitigated, the deleterious effects of TcdA on the monolayer's integrity. These events were not accompanied by alterations in ZO-1 and occludin protein levels; however, immunofluorescence microscopy revealed that both AOS and COS prevented the TcdA-induced occludin mislocalization. Finally, both NDOs accelerated TJ reassembly upon a calcium-switch assay. Overall, this study established the antipathogenic and barrier-protective capacity of AOS and COS against C. difficile and its toxin, TcdA, while revealing their ability to promote TJ reassembly in Caco-2 cells.
Collapse
Affiliation(s)
- Maria Eleni Mavrogeni
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Mostafa Asadpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Jo H Judernatz
- Structural Biochemistry Group, Bijvoet Centre for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Ingrid van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Marc M S M Wösten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Karin Strijbis
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Roland J Pieters
- Division of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
11
|
Tyeb S, Verma V, Kumar N. Polysaccharide based transdermal patches for chronic wound healing: Recent advances and clinical perspective. Carbohydr Polym 2023; 316:121038. [PMID: 37321732 DOI: 10.1016/j.carbpol.2023.121038] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/02/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Polysaccharides form a major class of natural polymers with diverse applications in biomedical science and tissue engineering. One of the key thrust areas for polysaccharide materials is skin tissue engineering and regeneration, whose market is estimated to reach around 31 billion USD globally by 2030, with a compounded annual growth rate of 10.46 %. Out of this, chronic wound healing and management is a major concern, especially for underdeveloped and developing nations, mainly due to poor access to medical interventions for such societies. Polysaccharide materials have shown promising results and clinical potential in recent decades with regard to chronic wound healing. Their low cost, ease of fabrication, biodegradability, and ability to form hydrogels make them ideal candidates for managing and healing such difficult-to-heal wounds. The present review presents a summary of the recently explored polysaccharide-based transdermal patches for managing and healing chronic wounds. Their efficacy and potency of healing both as active and passive wound dressings are evaluated in several in-vitro and in-vivo models. Finally, their clinical performances and future challenges are summarized to draw a road map towards their role in advanced wound care.
Collapse
Affiliation(s)
- Suhela Tyeb
- Department of Materials Engineering, Indian Institute of Science Bangalore, Bengaluru 560012, India
| | - Vivek Verma
- Department of Materials Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India; Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India; Samtel Centre for Display Technologies, Indian Institute of Technology Kanpur, Kanpur 208016, India; National Centre for Flexible Electronics, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Nitesh Kumar
- Department of Materials Engineering, Indian Institute of Technology Jammu, Jammu 181221, India.
| |
Collapse
|
12
|
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
|
13
|
Rouillard KR, Esther CP, Kissner WJ, Plott LM, Bowman DW, Markovetz MR, Hill DB. Combination Treatment to Improve Mucociliary Transport of Pseudomonas aeruginosa Biofilms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553173. [PMID: 37645913 PMCID: PMC10461968 DOI: 10.1101/2023.08.14.553173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
People with muco-obstructive pulmonary diseases such as cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) often have acute or chronic respiratory infections that are difficult to treat due in part to the accumulation of hyperconcentrated mucus within the airway. Mucus accumulation and obstruction promote chronic inflammation and infection and reduce therapeutic efficacy. Bacterial aggregates in the form of biofilms exhibit increased resistance to mechanical stressors from the immune response (e.g., phagocytosis) and chemical treatments including antibiotics. Herein, combination treatments designed to disrupt the mechanical properties of biofilms and potentiate antibiotic efficacy are investigated against mucus-grown Pseudomonas aeruginosa biofilms and optimized to 1) alter biofilm viscoelastic properties, 2) increase mucociliary transport rates, and 3) reduce bacterial viability. A disulfide bond reducing agent (tris(2-carboxyethyl)phosphine, TCEP), a surfactant (NP40), a biopolymer (hyaluronic acid, HA), a DNA degradation enzyme (DNase), and an antibiotic (tobramycin) are tested in various combinations to maximize biofilm disruption. The viscoelastic properties of biofilms are quantified with particle tracking microrheology and transport rates are quantified in a mucociliary transport device comprised of fully differentiated primary human bronchial epithelial cells. The combination of the NP40 with hyaluronic acid and tobramycin was the most effective at increasing mucociliary transport rates, decreasing the viscoelastic properties of mucus, and reducing bacterial viability. Multimechanistic targeting of biofilm infections may ultimately result in improved clinical outcomes, and the results of this study may be translated into future in vivo infection models.
Collapse
Affiliation(s)
| | | | | | - Lucas M Plott
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC 27599
| | - Dean W Bowman
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC 27599
| | | | - David B Hill
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC 27599
- Joint Department of Biomedical Engineering, UNC Chapel Hill, NC 27599
| |
Collapse
|
14
|
Chung J, Eisha S, Park S, Morris AJ, Martin I. How Three Self-Secreted Biofilm Exopolysaccharides of Pseudomonas aeruginosa, Psl, Pel, and Alginate, Can Each Be Exploited for Antibiotic Adjuvant Effects in Cystic Fibrosis Lung Infection. Int J Mol Sci 2023; 24:ijms24108709. [PMID: 37240055 DOI: 10.3390/ijms24108709] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
In cystic fibrosis (CF), pulmonary infection with Pseudomonas aeruginosa is a cause of increased morbidity and mortality, especially in patients for whom infection becomes chronic and there is reliance on long-term suppressive therapies. Current antimicrobials, though varied mechanistically and by mode of delivery, are inadequate not only due to their failure to eradicate infection but also because they do not halt the progression of lung function decline over time. One of the reasons for this failure is thought to be the biofilm mode of growth of P. aeruginosa, wherein self-secreted exopolysaccharides (EPSs) provide physical protection against antibiotics and an array of niches with resulting metabolic and phenotypic heterogeneity. The three biofilm-associated EPSs secreted by P. aeruginosa (alginate, Psl, and Pel) are each under investigation and are being exploited in ways that potentiate antibiotics. In this review, we describe the development and structure of P. aeruginosa biofilms before examining each EPS as a potential therapeutic target for combating pulmonary infection with P. aeruginosa in CF, with a particular focus on the current evidence for these emerging therapies and barriers to bringing these therapies into clinic.
Collapse
Affiliation(s)
- Jonathan Chung
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Shafinaz Eisha
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Subin Park
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Amanda J Morris
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Isaac Martin
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
15
|
Mechanisms and technology of marine oligosaccharides to control postharvest disease of fruits. Food Chem 2023; 404:134664. [DOI: 10.1016/j.foodchem.2022.134664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/18/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
|
16
|
The Role of Quorum Sensing Molecules in Bacterial-Plant Interactions. Metabolites 2023; 13:metabo13010114. [PMID: 36677039 PMCID: PMC9863971 DOI: 10.3390/metabo13010114] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/03/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Quorum sensing (QS) is a system of communication of bacterial cells by means of chemical signals called autoinducers, which modulate the behavior of entire populations of Gram-negative and Gram-positive bacteria. Three classes of signaling molecules have been recognized, Al-1, Al-2, Al-3, whose functions are slightly different. However, the phenomenon of quorum sensing is not only concerned with the interactions between bacteria, but the whole spectrum of interspecies interactions. A growing number of research results confirm the important role of QS molecules in the growth stimulation and defense responses in plants. Although many of the details concerning the signaling metabolites of the rhizosphere microflora and plant host are still unknown, Al-1 compounds should be considered as important components of bacterial-plant interactions, leading to the stimulation of plant growth and the biological control of phytopathogens. The use of class 1 autoinducers in plants to induce beneficial activity may be a practical solution to improve plant productivity under field conditions. In addition, researchers are also interested in tools that offer the possibility of regulating the activity of autoinducers by means of degrading enzymes or specific inhibitors (QSI). Current knowledge of QS and QSI provides an excellent foundation for the application of research to biopreparations in agriculture, containing a consortia of AHL-producing bacteria and QS inhibitors and limiting the growth of phytopathogenic organisms.
Collapse
|
17
|
Powell LC, Adams JYM, Quoraishi S, Py C, Oger A, Gazze SA, Francis LW, von Ruhland C, Owens D, Rye PD, Hill KE, Pritchard MF, Thomas DW. Alginate oligosaccharides enhance the antifungal activity of nystatin against candidal biofilms. Front Cell Infect Microbiol 2023; 13:1122340. [PMID: 36798083 PMCID: PMC9927220 DOI: 10.3389/fcimb.2023.1122340] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Background The increasing prevalence of invasive fungal infections in immuno-compromised patients is a considerable cause of morbidity and mortality. With the rapid emergence of antifungal resistance and an inadequate pipeline of new therapies, novel treatment strategies are now urgently required. Methods The antifungal activity of the alginate oligosaccharide OligoG in conjunction with nystatin was tested against a range of Candida spp. (C. albicans, C. glabrata, C. parapsilosis, C. auris, C. tropicalis and C. dubliniensis), in both planktonic and biofilm assays, to determine its potential clinical utility to enhance the treatment of candidal infections. The effect of OligoG (0-6%) ± nystatin on Candida spp. was examined in minimum inhibitory concentration (MIC) and growth curve assays. Antifungal effects of OligoG and nystatin treatment on biofilm formation and disruption were characterized using confocal laser scanning microscopy (CLSM), scanning electron microscopy (SEM) and ATP cellular viability assays. Effects on the cell membrane were determined using permeability assays and transmission electron microscopy (TEM). Results MIC and growth curve assays demonstrated the synergistic effects of OligoG (0-6%) with nystatin, resulting in an up to 32-fold reduction in MIC, and a significant reduction in the growth of C. parapsilosis and C. auris (minimum significant difference = 0.2 and 0.12 respectively). CLSM and SEM imaging demonstrated that the combination treatment of OligoG (4%) with nystatin (1 µg/ml) resulted in significant inhibition of candidal biofilm formation on glass and clinical grade silicone surfaces (p < 0.001), with increased cell death (p < 0.0001). The ATP biofilm disruption assay demonstrated a significant reduction in cell viability with OligoG (4%) alone and the combined OligoG/nystatin (MIC value) treatment (p < 0.04) for all Candida strains tested. TEM studies revealed the combined OligoG/nystatin treatment induced structural reorganization of the Candida cell membrane, with increased permeability when compared to the untreated control (p < 0.001). Conclusions Antimicrobial synergy between OligoG and nystatin against Candida spp. highlights the potential utility of this combination therapy in the prevention and topical treatment of candidal biofilm infections, to overcome the inherent tolerance of biofilm structures to antifungal agents.
Collapse
Affiliation(s)
- Lydia C. Powell
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- Microbiology and Infectious Disease group, Swansea University Medical School, Swansea, United Kingdom
- *Correspondence: Lydia C. Powell,
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Sadik Quoraishi
- Otolaryngology Department, New Cross Hospital, Wolverhampton, United Kingdom
| | - Charlène Py
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Anaϊs Oger
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Salvatore A. Gazze
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis W. Francis
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Christopher von Ruhland
- Central Biotechnology Services, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David Owens
- Head and Neck Directorate, University Hospital of Wales, Cardiff, United Kingdom
| | | | - Katja E. Hill
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Manon F. Pritchard
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - David W. Thomas
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| |
Collapse
|
18
|
Zhang C, Li M, Rauf A, Khalil AA, Shan Z, Chen C, Rengasamy KRR, Wan C. Process and applications of alginate oligosaccharides with emphasis on health beneficial perspectives. Crit Rev Food Sci Nutr 2023; 63:303-329. [PMID: 34254536 DOI: 10.1080/10408398.2021.1946008] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alginates are linear polymers comprising 40% of the dry weight of algae possess various applications in food and biomedical industries. Alginate oligosaccharides (AOS), a degradation product of alginate, is now gaining much attention for their beneficial role in food, pharmaceutical and agricultural industries. Hence this review was aimed to compile the information on alginate and AOS (prepared from seaweeds) during 1994-2020. As per our knowledge, this is the first review on the potential use of alginate oligosaccharides in different fields. The alginate derivatives are grouped according to their applications. They are involved in the isolation process and show antimicrobial, antioxidant, anti-inflammatory, antihypertension, anticancer, and immunostimulatory properties. AOS also have significant applications in prebiotics, nutritional supplements, plant growth development and others products.
Collapse
Affiliation(s)
- Chunhua Zhang
- College of Agriculture and Forestry, Pu'er University, Pu'er, Yunnan, China
| | - Mingxi Li
- Research Center of Tea and Tea Culture, College of Agronomy, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Khyber Pakhtunkhwa (KP), Pakistan
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Diet and Nutritional Sciences, The University of Lahore, Lahore, Pakistan
| | - Zhiguo Shan
- College of Agriculture and Forestry, Pu'er University, Pu'er, Yunnan, China
| | - Chuying Chen
- Research Center of Tea and Tea Culture, College of Agronomy, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Kannan R R Rengasamy
- Green Biotechnologies Research Centre of Excellence, University of Limpopo, Polokwane, Sovenga, South Africa
| | - Chunpeng Wan
- Research Center of Tea and Tea Culture, College of Agronomy, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
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
|
20
|
Alginates Combined with Natural Polymers as Valuable Drug Delivery Platforms. Mar Drugs 2022; 21:md21010011. [PMID: 36662184 PMCID: PMC9861938 DOI: 10.3390/md21010011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Alginates (ALG) have been used in biomedical and pharmaceutical technologies for decades. ALG are natural polymers occurring in brown algae and feature multiple advantages, including biocompatibility, low toxicity and mucoadhesiveness. Moreover, ALG demonstrate biological activities per se, including anti-hyperlipidemic, antimicrobial, anti-reflux, immunomodulatory or anti-inflammatory activities. ALG are characterized by gelling ability, one of the most frequently utilized properties in the drug form design. ALG have numerous applications in pharmaceutical technology that include micro- and nanoparticles, tablets, mucoadhesive dosage forms, wound dressings and films. However, there are some shortcomings, which impede the development of modified-release dosage forms or formulations with adequate mechanical strength based on pure ALG. Other natural polymers combined with ALG create great potential as drug carriers, improving limitations of ALG matrices. Therefore, in this paper, ALG blends with pectins, chitosan, gelatin, and carrageenans were critically reviewed.
Collapse
|
21
|
Cao S, Li L, Zhu B, Yao Z. Alginate modifying enzymes: An updated comprehensive review of the mannuronan C5-epimerases. ALGAL RES 2022. [DOI: 10.1016/j.algal.2022.102952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
Bhowmik A, Chunhavacharatorn P, Bhargav S, Malhotra A, Sendrayakannan A, Kharkar PS, Nirmal NP, Chauhan A. Human Milk Oligosaccharides as Potential Antibiofilm Agents: A Review. Nutrients 2022; 14:nu14235112. [PMID: 36501142 PMCID: PMC9737902 DOI: 10.3390/nu14235112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 12/04/2022] Open
Abstract
Surface-associated bacterial communities called biofilms are ubiquitous in nature. Biofilms are detrimental in medical settings due to their high tolerance to antibiotics and may alter the final pathophysiological outcome of many healthcare-related infections. Several innovative prophylactic and therapeutic strategies targeting specific mechanisms and/or pathways have been discovered and exploited in the clinic. One such emerging and original approach to dealing with biofilms is the use of human milk oligosaccharides (HMOs), which are the third most abundant solid component in human milk after lactose and lipids. HMOs are safe to consume (GRAS status) and act as prebiotics by inducing the growth and colonization of gut microbiota, in addition to strengthening the intestinal epithelial barrier, thereby protecting from pathogens. Moreover, HMOs can disrupt biofilm formation and inhibit the growth of specific microbes. In the present review, we summarize the potential of HMOs as antibacterial and antibiofilm agents and, hence, propose further investigations on using HMOs for new-age therapeutic interventions.
Collapse
Affiliation(s)
- Ankurita Bhowmik
- Department of Microbiology, Tripura University, Agartala 799022, India
| | | | - Sharanya Bhargav
- Department of Molecular Biology, Yuvaraja’s College, Mysuru 570005, India
| | - Akshit Malhotra
- Department of Microbiology, Tripura University, Agartala 799022, India
- Invisiobiome, New Delhi 110066, India
| | - Akalya Sendrayakannan
- Department of Food Engineering and Technology, Institute of Chemical Technology (ICT), Nathalal Parekh Marg, Matunga, Mumbai 400019, India
| | - Prashant S. Kharkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology (ICT), Nathalal Parekh Marg, Matunga, Mumbai 400019, India
- Correspondence: (P.S.K.); (N.P.N.); (A.C.)
| | - Nilesh Prakash Nirmal
- Institute of Nutrition, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
- Correspondence: (P.S.K.); (N.P.N.); (A.C.)
| | - Ashwini Chauhan
- Department of Microbiology, Tripura University, Agartala 799022, India
- Correspondence: (P.S.K.); (N.P.N.); (A.C.)
| |
Collapse
|
23
|
Zhou L, Meng Q, Zhang R, Jiang B, Liu X, Chen J, Zhang T. Characterization of a Novel Polysaccharide Lyase Family 5 Alginate Lyase with PolyM Substrate Specificity. Foods 2022; 11:3527. [PMID: 36360141 PMCID: PMC9655155 DOI: 10.3390/foods11213527] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 09/19/2023] Open
Abstract
Alginate lyases (ALyases) have been widely applied in enzymatically degrading alginate for the preparation of alginate oligosaccharides (AOS), which possess a range of excellent physiological benefits including immunoregulatory, antivirus, and antidiabetic properties. Among the characterized ALyases, the number of ALyases with strict substrate specificity which possess potential in directed preparation of AOS is quite small. ALyases of polysaccharides lyase (PL) 5 family have been reported to perform poly-β-D-mannuronic acid (Poly-M) substrate specificity. However, there have been fewer studies with a comprehensive characterization and comparison of PL 5 family ALyases. In this study, a putative PL 5 family ALyase PMD was cloned from Pseudomonas mendocina and expressed in Escherichia coli. The novel ALyase presented maximum activity at 30 °C and pH 7.0. PMD displayed pH stability properties under the range of pH 5 to pH 9, which retained more than 80% relative activity, even when incubated for 48 h. Product analysis indicated that PMD might be an endolytic ALyase with strict Poly M substrate specificity and yield disaccharide and trisaccharide as main products. In addition, residues K58, R66, Y248, and R344 were proposed to be the potential key residues for catalysis via site-directed mutation. Detailed characterization of PMD and comprehensive comparisons could supply some different information about properties of PL 5 ALyases which might be helpful for its application in the directed production of AOS.
Collapse
Affiliation(s)
- Licheng Zhou
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Qing Meng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Ran Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Bo Jiang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Xiaoyong Liu
- Shandong Haizhibao Ocean Technology Co., Ltd., Weihai 264333, China
| | - Jingjing Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
24
|
Hills OJ, Yong CW, Scott AJ, Smith J, Chappell HF. Polyguluronate simulations shed light onto the therapeutic action of OligoG CF-5/20. Bioorg Med Chem 2022; 72:116945. [PMID: 36037625 DOI: 10.1016/j.bmc.2022.116945] [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: 05/30/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/25/2022]
Abstract
Chronic mucoid P. aeruginosa cystic fibrosis (CF) lung infections are associated with the development of a biofilm composed of anionic acetylated exopolysaccharide (EPS) alginate, electrostatically stabilised by extracellular Ca2+ ions. OligoG CF-5/20, a low molecular weight guluronate rich oligomer, is emerging as a novel therapeutic capable of disrupting mature P. aeruginosa biofilms. However, its method of therapeutic action on the mucoid biofilm EPS is not definitively known at a molecular level. This work, utilising molecular dynamics (MD) and Density-Functional Theory (DFT), has revealed that OligoG CF-5/20 interaction with the EPS is facilitated solely through bridging Ca2+ ions, which are not liberated from their native EPS binding sites upon OligoG CF-5/20 dispersal, suggesting that OligoG CF-5/20 does not cause disruptions to mature P. aeruginosa biofilms through breaking EPS-Ca2+-EPS ionic cross-links. Rather it is likely that the therapeutic activity arises from sequestering free Ca2+ ions and preventing further Ca2+ induced EPS aggregation.
Collapse
Affiliation(s)
- Oliver J Hills
- School of Food Science & Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| | - Chin W Yong
- Scientific Computing Department, Science and Technology Facilities Council, Daresbury Laboratory, Keckwick Lane, Daresbury, Warrington, WA4 4AD, UK; Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Andrew J Scott
- School of Chemical & Process Engineering, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| | - James Smith
- School of Food Science & Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| | - Helen F Chappell
- School of Food Science & Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK.
| |
Collapse
|
25
|
Powell LC, Cullen JK, Boyle GM, De Ridder T, Yap PY, Xue W, Pierce CJ, Pritchard MF, Menzies GE, Abdulkarim M, Adams JYM, Stokniene J, Francis LW, Gumbleton M, Johns J, Hill KE, Jones AV, Parsons PG, Reddell P, Thomas DW. Topical, immunomodulatory epoxy-tiglianes induce biofilm disruption and healing in acute and chronic skin wounds. Sci Transl Med 2022; 14:eabn3758. [DOI: 10.1126/scitranslmed.abn3758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The management of antibiotic-resistant, bacterial biofilm infections in chronic skin wounds is an increasing clinical challenge. Despite advances in diagnosis, many patients do not derive benefit from current anti-infective/antibiotic therapies. Here, we report a novel class of naturally occurring and semisynthetic epoxy-tiglianes, derived from the Queensland blushwood tree (
Fontainea picrosperma)
, and demonstrate their antimicrobial activity (modifying bacterial growth and inducing biofilm disruption), with structure/activity relationships established against important human pathogens. In vitro, the lead candidate EBC-1013 stimulated protein kinase C (PKC)–dependent neutrophil reactive oxygen species (ROS) induction and NETosis and increased expression of wound healing–associated cytokines, chemokines, and antimicrobial peptides in keratinocytes and fibroblasts. In vivo, topical EBC-1013 induced rapid resolution of infection with increased matrix remodeling in acute thermal injuries in calves. In chronically infected diabetic mouse wounds, treatment induced cytokine/chemokine production, inflammatory cell recruitment, and complete healing (in six of seven wounds) with ordered keratinocyte differentiation. These results highlight a nonantibiotic approach involving contrasting, orthogonal mechanisms of action combining targeted biofilm disruption and innate immune induction in the treatment of chronic wounds.
Collapse
Affiliation(s)
- Lydia C. Powell
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
- Centre for Nanohealth, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Jason K. Cullen
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Glen M. Boyle
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Tom De Ridder
- QBiotics Group Limited Yungaburra, Queensland 4884, Australia
| | - Pei-Yi Yap
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Wenya Xue
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | - Carly J. Pierce
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Manon F. Pritchard
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | | | - Muthanna Abdulkarim
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, UK
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | - Joana Stokniene
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | - Lewis W. Francis
- Centre for Nanohealth, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, UK
| | - Jenny Johns
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Katja E. Hill
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | - Adam V. Jones
- Oral Pathology, Cardiff and Vale University Health Board , Cardiff CF14 4XY, UK
| | - Peter G. Parsons
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Paul Reddell
- QBiotics Group Limited Yungaburra, Queensland 4884, Australia
| | - David W. Thomas
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| |
Collapse
|
26
|
Attenuation of Pseudomonas aeruginosa Quorum Sensing Virulence of Biofilm and Pyocyanin by mBTL-Loaded Calcium Alginate Nanoparticles. Polymers (Basel) 2022; 14:polym14173655. [PMID: 36080730 PMCID: PMC9459901 DOI: 10.3390/polym14173655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/27/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Pseudomonas aeruginosa contributes to many chronic infections and has been found to be resistant to multiple antibiotics. Pseudomonas use a quorum sensing system (QS) to control biofilm establishment and virulence factors, and, thus, quorum sensing inhibitors (QSIs), such as meta-bromo-thiolactone (mBTL), are promising anti-infective agents. Accordingly, this study intended to investigate the antibacterial and anti-virulence activity of mBTL-loaded calcium alginate nanoparticles (CANPs) against Pseudomonas aeruginosa and different QS mutants. The results show that the mBTL-CANPs had higher antibacterial activity, which was made evident by decreases in all tested strains except the ∆lasR/∆rhlR double mutant, with MIC50 (0.5 mg/mL) of mBTL-CANPs compared with free mBTL at MIC50 (˃1 mg/mL). The biofilm formation of P. aeruginosa and some QS-deficient mutants were reduced in response to 0.5-0.125 mg/mL of mBTL-encapsulating CANPs. The pyocyanin production of the tested strains except ∆lasA and ∆rhlR decreased when challenged with 0.5 mg/mL of mBTL-loaded NPs. The subsequent characterization of the cytotoxic effect of these NPs on human lung epithelial cells (A549) and cystic fibrosis fibroblast cells (LL 29) demonstrated that synthesized NPs were cytocompatible at MIC50 in both cell lines and markedly reduced the cytotoxic effect observed with mBTL alone on these cells. The resulting formulation reduced the P. aeruginosa strains' adhesion to A549 comparably with mBTL, suggesting their potential anti-adhesive effect. Given the virulence suppressing action, cytocompatibility, and enhanced anti-biofilm effect of mBTL-CANPs, and the advantage of alginate-based NPs as an antimicrobial delivery system these nanoparticles have great potential in the prophylaxis and treatment of infection caused by Pseudomonas aeruginosa.
Collapse
|
27
|
Hazime N, Belguesmia Y, Kempf I, Barras A, Drider D, Boukherroub R. Enhancing Colistin Activity against Colistin-Resistant Escherichia coli through Combination with Alginate Nanoparticles and Small Molecules. Pharmaceuticals (Basel) 2022; 15:ph15060682. [PMID: 35745601 PMCID: PMC9227550 DOI: 10.3390/ph15060682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
Bacterial resistance to antibiotics has become a major public health problem worldwide, with the yearly number of deaths exceeding 700,000. To face this well-acknowledged threat, new molecules and therapeutic methods are considered. In this context, the application of nanotechnology to fight bacterial infection represents a viable approach and has experienced tremendous developments in the last decades. Escherichia coli (E. coli) is responsible for severe diarrhea, notably in the breeding sector, and especially in pig farming. The resulting infection (named colibacillosis) occurs in young piglets and could lead to important economic losses. Here, we report the design of several new formulations based on colistin loaded on alginate nanoparticles (Alg NPs) in the absence, but also in the presence, of small molecules, such as components of essential oils, polyamines, and lactic acid. These new formulations, which are made by concomitantly binding colistin and small molecules to Alg NPs, were successfully tested against E. coli 184, a strain resistant to colistin. When colistin was associated with Alg NPs, the minimal inhibition concentration (MIC) decreased from 8 to 1 µg/mL. It is notable that when menthol or lactic acid was co-loaded with colistin on Alg NPs, the MIC of colistin drastically decreased, reaching 0.31 or 0.62 µg/mL, respectively. These novel bactericidal formulations, whose innocuity towards eukaryotic HT-29 cells was established in vitro, are presumed to permeabilize the bacterial membrane and provoke the leakage of intracellular proteins. Our findings revealed the potentiating effect of the Alg NPs on colistin, but also of the small molecules mentioned above. Such ecological and economical formulations are easy to produce and could be proposed, after confirmation by in vivo and toxicology tests, as therapeutic strategies to replace fading antibiotics.
Collapse
Affiliation(s)
- Noura Hazime
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France; (N.H.); (A.B.)
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D’Opale, ICV-Institut Charles Viollette, 59000 Lille, France; (Y.B.); (D.D.)
| | - Yanath Belguesmia
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D’Opale, ICV-Institut Charles Viollette, 59000 Lille, France; (Y.B.); (D.D.)
| | - Isabelle Kempf
- Agence Nationale de Sécurité Sanitaire de L'Alimentation, de L'Environnement et du Travail, Laboratoire de Ploufragan-Plouzané-Niort, Unité Mycoplasmologie Bactériologie Antibiorésistance, 22440 Ploufragan, France;
| | - Alexandre Barras
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France; (N.H.); (A.B.)
| | - Djamel Drider
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D’Opale, ICV-Institut Charles Viollette, 59000 Lille, France; (Y.B.); (D.D.)
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France; (N.H.); (A.B.)
- Correspondence:
| |
Collapse
|
28
|
Barzkar N, Sheng R, Sohail M, Jahromi ST, Babich O, Sukhikh S, Nahavandi R. Alginate Lyases from Marine Bacteria: An Enzyme Ocean for Sustainable Future. Molecules 2022; 27:3375. [PMID: 35684316 PMCID: PMC9181867 DOI: 10.3390/molecules27113375] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 12/13/2022] Open
Abstract
The cell wall of brown algae contains alginate as a major constituent. This anionic polymer is a composite of β-d-mannuronate (M) and α-l-guluronate (G). Alginate can be degraded into oligosaccharides; both the polymer and its products exhibit antioxidative, antimicrobial, and immunomodulatory activities and, hence, find many commercial applications. Alginate is attacked by various enzymes, collectively termed alginate lyases, that degrade glycosidic bonds through β-elimination. Considering the abundance of brown algae in marine ecosystems, alginate is an important source of nutrients for marine organisms, and therefore, alginate lyases play a significant role in marine carbon recycling. Various marine microorganisms, particularly those that thrive in association with brown algae, have been reported as producers of alginate lyases. Conceivably, the marine-derived alginate lyases demonstrate salt tolerance, and many are activated in the presence of salts and, therefore, find applications in the food industry. Therefore, this review summarizes the structural and biochemical features of marine bacterial alginate lyases along with their applications. This comprehensive information can aid in the expansion of future prospects of alginate lyases.
Collapse
Affiliation(s)
- Noora Barzkar
- Department of Marine Biology, Faculty of Marine Science and Technology, University of Hormozgan, Bandar Abbas 3995, Iran
| | - Ruilong Sheng
- CQM—Centro de Química da Madeira, Campus da Penteada, Universidade da Madeira, 9000-390 Funchal, Portugal;
- Department of Radiology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Muhammad Sohail
- Department of Microbiology, University of Karachi, Karachi 75270, Pakistan;
| | - Saeid Tamadoni Jahromi
- Persian Gulf and Oman Sea Ecology Research Center, Iranian Fisheries Sciences Research Institute, Agricultural Research Education and Extension Organization (AREEO), Bandar Abbas 9145, Iran;
| | - Olga Babich
- Institute of Living Systems, Immanuel Kant Baltic Federal University, A. Nevskogo Street 14, Kaliningrad 236016, Russia; (O.B.); (S.S.)
| | - Stanislav Sukhikh
- Institute of Living Systems, Immanuel Kant Baltic Federal University, A. Nevskogo Street 14, Kaliningrad 236016, Russia; (O.B.); (S.S.)
| | - Reza Nahavandi
- Animal Science Research Institute of Iran (ASRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj 8361, Iran;
| |
Collapse
|
29
|
Lu S, Na K, Wei J, Zhang L, Guo X. Alginate oligosaccharides: The structure-function relationships and the directional preparation for application. Carbohydr Polym 2022; 284:119225. [PMID: 35287920 DOI: 10.1016/j.carbpol.2022.119225] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/20/2022] [Accepted: 02/02/2022] [Indexed: 01/02/2023]
Abstract
Alginate oligosaccharides (AOS) are degradation products of alginate extracted from brown algae. With low molecular weight, high water solubility, and good biological activity, AOS present anti-inflammatory, antimicrobial, antioxidant, and antitumor properties. They also exert growth-promoting effects in animals and plants. Three types of AOS, mannuronate oligosaccharides (MAOS), guluronate oligosaccharides (GAOS), and heterozygous mannuronate and guluronate oligosaccharides (HAOS), can be produced from alginate by enzymatic hydrolysis. Thus far, most studies on the applications and biological activities of AOS have been based mainly on a hybrid form of HAOS. To improve the directional production of AOS for practical applications, systematic studies on the structures and related biological activities of AOS are needed. This review provides a summary of current understanding of structure-function relationships and advances in the production of AOS. The current challenges and opportunities in the application of AOS is suggested to guide the precise application of AOS in practice.
Collapse
Affiliation(s)
- Shuang Lu
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China
| | - Kai Na
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China
| | - Jiani Wei
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China
| | - Li Zhang
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China
| | - Xiaohua Guo
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China.
| |
Collapse
|
30
|
Bi D, Yang X, Lu J, Xu X. Preparation and potential applications of alginate oligosaccharides. Crit Rev Food Sci Nutr 2022; 63:10130-10147. [PMID: 35471191 DOI: 10.1080/10408398.2022.2067832] [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] [Indexed: 12/11/2022]
Abstract
Alginate, a linear polymer consisting of β-D-mannuronic acid (M) and α-L-guluronic acid (G) with 1,4-glycosidic linkages and comprising 40% of the dry weight of algae, possesses various applications in the food and nutraceutical industries. However, the potential applications of alginate are restricted in some fields because of its low water solubility and high solution viscosity. Alginate oligosaccharides (AOS) on the other hand, have low molecular weight which result in better water solubility. Hence, it becomes a more popular target to be researched in recent years for its use in foods and nutraceuticals. AOS can be obtained by multiple degradation methods, including enzymatic degradation, from alginate or alginate-derived poly G and poly M. AOS have unique bioactivity and can bring human health benefits, which render them potentials to be developed/incorporated into functional food. This review comprehensively covers methods of the preparation and analysis of AOS, and discussed the potential applications of AOS in foods and nutraceuticals.
Collapse
Affiliation(s)
- Decheng Bi
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, and Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, PR China
- School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Xu Yang
- School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Jun Lu
- School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
- School of Public Health and Interdisciplinary Studies, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Xu Xu
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, and Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, PR China
| |
Collapse
|
31
|
Alginate oligosaccharides enhance diffusion and activity of colistin in a mucin-rich environment. Sci Rep 2022; 12:4986. [PMID: 35322119 PMCID: PMC8943044 DOI: 10.1038/s41598-022-08927-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
In a number of chronic respiratory diseases e.g. cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD), the production of viscous mucin reduces pulmonary function and represents an effective barrier to diffusion of inhaled therapies e.g. antibiotics. Here, a 2-compartment Transwell model was developed to study impaired diffusion of the antibiotic colistin across an artificial sputum (AS) matrix/medium and to quantify its antimicrobial activity against Pseudomonas aeruginosa NH57388A biofilms (alone and in combination with mucolytic therapy). High-performance liquid chromatography coupled with fluorescence detection (HPLC-FLD) revealed that the presence of AS medium significantly reduced the rate of colistin diffusion (> 85% at 48 h; p < 0.05). Addition of alginate oligosaccharide (OligoG CF-5/20) significantly improved colistin diffusion by 3.7 times through mucin-rich AS medium (at 48 h; p < 0.05). Increased diffusion of colistin with OligoG CF-5/20 was shown (using confocal laser scanning microscopy and COMSTAT image analysis) to be associated with significantly increased bacterial killing (p < 0.05). These data support the use of this model to study drug and small molecule delivery across clinically-relevant diffusion barriers. The findings indicate the significant loss of colistin and reduced effectiveness that occurs with mucin binding, and support the use of mucolytics to improve antimicrobial efficacy and lower antibiotic exposure.
Collapse
|
32
|
Evaluating the alginate oligosaccharide (OligoG) as a therapy for Burkholderia cepacia complex cystic fibrosis lung infection. J Cyst Fibros 2022; 21:821-829. [DOI: 10.1016/j.jcf.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/15/2021] [Accepted: 01/09/2022] [Indexed: 11/15/2022]
|
33
|
Pseudomonas aeruginosa in the Cystic Fibrosis Lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:347-369. [DOI: 10.1007/978-3-031-08491-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Rosiak P, Latanska I, Paul P, Sujka W, Kolesinska B. Modification of Alginates to Modulate Their Physic-Chemical Properties and Obtain Biomaterials with Different Functional Properties. Molecules 2021; 26:7264. [PMID: 34885846 PMCID: PMC8659150 DOI: 10.3390/molecules26237264] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 01/02/2023] Open
Abstract
Modified alginates have a wide range of applications, including in the manufacture of dressings and scaffolds used for regenerative medicine, in systems for selective drug delivery, and as hydrogel materials. This literature review discusses the methods used to modify alginates and obtain materials with new or improved functional properties. It discusses the diverse biological and functional activity of alginates. It presents methods of modification that utilize both natural and synthetic peptides, and describes their influence on the biological properties of the alginates. The success of functionalization depends on the reaction conditions being sufficient to guarantee the desired transformations and provide modified alginates with new desirable properties, but mild enough to prevent degradation of the alginates. This review is a literature description of efficient methods of alginate functionalization using biologically active ligands. Particular attention was paid to methods of alginate functionalization with peptides, because the combination of the properties of alginates and peptides leads to the obtaining of conjugates with properties resulting from both components as well as a completely new, different functionality.
Collapse
Affiliation(s)
- Piotr Rosiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Ilona Latanska
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Paulina Paul
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Witold Sujka
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Beata Kolesinska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| |
Collapse
|
35
|
Gao SK, Yin R, Wang XC, Jiang HN, Liu XX, Lv W, Ma Y, Zhou YX. Structure Characteristics, Biochemical Properties, and Pharmaceutical Applications of Alginate Lyases. Mar Drugs 2021; 19:628. [PMID: 34822499 PMCID: PMC8618178 DOI: 10.3390/md19110628] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/27/2021] [Accepted: 11/06/2021] [Indexed: 02/07/2023] Open
Abstract
Alginate, the most abundant polysaccharides of brown algae, consists of various proportions of uronic acid epimers α-L-guluronic acid (G) and β-D-mannuronic acid (M). Alginate oligosaccharides (AOs), the degradation products of alginates, exhibit excellent bioactivities and a great potential for broad applications in pharmaceutical fields. Alginate lyases can degrade alginate to functional AOs with unsaturated bonds or monosaccharides, which can facilitate the biorefinery of brown algae. On account of the increasing applications of AOs and biorefinery of brown algae, there is a scientific need to explore the important aspects of alginate lyase, such as catalytic mechanism, structure, and property. This review covers fundamental aspects and recent developments in basic information, structural characteristics, the structure-substrate specificity or catalytic efficiency relationship, property, molecular modification, and applications. To meet the needs of biorefinery systems of a broad array of biochemical products, alginate lyases with special properties, such as salt-activated, wide pH adaptation range, and cold adaptation are outlined. Withal, various challenges in alginate lyase research are traced out, and future directions, specifically on the molecular biology part of alginate lyases, are delineated to further widen the horizon of these exceptional alginate lyases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yan-Xia Zhou
- Marine College, Shandong University, Weihai 264209, China; (S.-K.G.); (R.Y.); (X.-C.W.); (H.-N.J.); (X.-X.L.); (W.L.); (Y.M.)
| |
Collapse
|
36
|
The Antibiofilm Nanosystems for Improved Infection Inhibition of Microbes in Skin. Molecules 2021; 26:molecules26216392. [PMID: 34770799 PMCID: PMC8587837 DOI: 10.3390/molecules26216392] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Biofilm formation is an important virulence factor for the opportunistic microorganisms that elicit skin infections. The recalcitrant feature of biofilms and their antibiotic tolerance impose a great challenge on the use of conventional therapies. Most antibacterial agents have difficulty penetrating the matrix produced by a biofilm. One novel approach to address these concerns is to prevent or inhibit the formation of biofilms using nanoparticles. The advantages of using nanosystems for antibiofilm applications include high drug loading efficiency, sustained or prolonged drug release, increased drug stability, improved bioavailability, close contact with bacteria, and enhanced accumulation or targeting to biomasses. Topically applied nanoparticles can act as a strategy for enhancing antibiotic delivery into the skin. Various types of nanoparticles, including metal oxide nanoparticles, polymeric nanoparticles, liposomes, and lipid-based nanoparticles, have been employed for topical delivery to treat biofilm infections on the skin. Moreover, nanoparticles can be designed to combine with external stimuli to produce magnetic, photothermal, or photodynamic effects to ablate the biofilm matrix. This study focuses on advanced antibiofilm approaches based on nanomedicine for treating skin infections. We provide in-depth descriptions on how the nanoparticles could effectively eliminate biofilms and any pathogens inside them. We then describe cases of using nanoparticles for antibiofilm treatment of the skin. Most of the studies included in this review were supported by in vivo animal infection models. This article offers an overview of the benefits of nanosystems for treating biofilms grown on the skin.
Collapse
|
37
|
Asadpoor M, Ithakisiou GN, van Putten JPM, Pieters RJ, Folkerts G, Braber S. Antimicrobial Activities of Alginate and Chitosan Oligosaccharides Against Staphylococcus aureus and Group B Streptococcus. Front Microbiol 2021; 12:700605. [PMID: 34589067 PMCID: PMC8473942 DOI: 10.3389/fmicb.2021.700605] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/17/2021] [Indexed: 12/22/2022] Open
Abstract
The bacterial pathogens Streptococcus agalactiae (GBS) and Staphylococcus aureus (S. aureus) cause serious infections in humans and animals. The emergence of antibiotic-resistant isolates and bacterial biofilm formation entails the urge of novel treatment strategies. Recently, there is a profound scientific interest in the capabilities of non-digestible oligosaccharides as antimicrobial and anti-biofilm agents as well as adjuvants in antibiotic combination therapies. In this study, we investigated the potential of alginate oligosaccharides (AOS) and chitosan oligosaccharides (COS) as alternative for, or in combination with antibiotic treatment. AOS (2-16%) significantly decreased GBS V growth by determining the minimum inhibitory concentration. Both AOS (8 and 16%) and COS (2-16%) were able to prevent biofilm formation by S. aureus wood 46. A checkerboard biofilm formation assay demonstrated a synergistic effect of COS and clindamycin on the S. aureus biofilm formation, while AOS (2 and 4%) were found to sensitize GBS V to trimethoprim. In conclusion, AOS and COS affect the growth of GBS V and S. aureus wood 46 and can function as anti-biofilm agents. The promising effects of AOS and COS in combination with different antibiotics may offer new opportunities to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Mostafa Asadpoor
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Georgia-Nefeli Ithakisiou
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Jos P. M. van Putten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Roland J. Pieters
- Division of Medicinal Chemistry and Chemical Biology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Saskia Braber
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
38
|
Tran HA, Tran PA. In Situ Coatings of Silver Nanoparticles for Biofilm Treatment in Implant-Retention Surgeries: Antimicrobial Activities in Monoculture and Coculture. ACS APPLIED MATERIALS & INTERFACES 2021; 13:41435-41444. [PMID: 34448395 DOI: 10.1021/acsami.1c08239] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Bacterial biofilms are indicated in most medical device-associated infections. Treating these biofilms is challenging yet critically important for applications such as in device-retention surgeries, which can have reinfection rates of up to 80%. This in vitro study centered around our new method of treating biofilm and preventing reinfection. Ionic silver (Ag, in the form of silver nitrate) combined with dopamine and a biofilm-lysing enzyme (α-amylase) were applied to model 4-day-old Staphylococcus aureus biofilms on titanium substrates to degrade the extracellular matrix of the biofilm and kill the biofilm bacteria. In this process, the oxidative self-polymerization of dopamine converted Ag ions into Ag nanoparticles that, together with the resultant self-adhering polydopamine (PDA), formed coatings that strongly bound to the treated substrates. Surprisingly, although these Ag/PDA coatings significantly reduced S. aureus growth in standard bacterial monoculture, they showed much lower antimicrobial activity in coculture of the bacteria and osteoblastic MC3T3-E1 cells in which the bacteria were also found attached to the osteoblasts. This S. aureus- osteoblast interaction was also linked to bacterial survival against gentamicin treatment observed in coculture. Our study thus provided clear evidence suggesting that bacteria's interactions with tissue cells surrounding implants may significantly contribute to their resistance to antimicrobial treatment.
Collapse
Affiliation(s)
- Hien A Tran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, QUT, 2 George Street, Brisbane, Queensland 4000, Australia
| | - Phong A Tran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, QUT, 2 George Street, Brisbane, Queensland 4000, Australia
| |
Collapse
|
39
|
Scoffone VC, Trespidi G, Barbieri G, Irudal S, Perrin E, Buroni S. Role of RND Efflux Pumps in Drug Resistance of Cystic Fibrosis Pathogens. Antibiotics (Basel) 2021; 10:863. [PMID: 34356783 PMCID: PMC8300704 DOI: 10.3390/antibiotics10070863] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/01/2021] [Accepted: 07/13/2021] [Indexed: 01/21/2023] Open
Abstract
Drug resistance represents a great concern among people with cystic fibrosis (CF), due to the recurrent and prolonged antibiotic therapy they should often undergo. Among Multi Drug Resistance (MDR) determinants, Resistance-Nodulation-cell Division (RND) efflux pumps have been reported as the main contributors, due to their ability to extrude a wide variety of molecules out of the bacterial cell. In this review, we summarize the principal RND efflux pump families described in CF pathogens, focusing on the main Gram-negative bacterial species (Pseudomonas aeruginosa, Burkholderia cenocepacia, Achromobacter xylosoxidans, Stenotrophomonas maltophilia) for which a predominant role of RND pumps has been associated to MDR phenotypes.
Collapse
Affiliation(s)
- Viola Camilla Scoffone
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (S.I.)
| | - Gabriele Trespidi
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (S.I.)
| | - Giulia Barbieri
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (S.I.)
| | - Samuele Irudal
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (S.I.)
| | - Elena Perrin
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Silvia Buroni
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.T.); (G.B.); (S.I.)
| |
Collapse
|
40
|
Wang M, Chen L, Zhang Z. Potential applications of alginate oligosaccharides for biomedicine - A mini review. Carbohydr Polym 2021; 271:118408. [PMID: 34364551 DOI: 10.1016/j.carbpol.2021.118408] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/23/2021] [Accepted: 07/03/2021] [Indexed: 01/02/2023]
Abstract
Extensive research on marine algae, especially on their health-promoting properties, has been conducted. Various ingredients with potential biomedical applications have been discovered and extracted from marine algae. Alginate oligosaccharides are low molecular weight alginate polysaccharides present in cell walls of brown algae. They exhibit various health benefits such as anti-inflammatory, anti-microbial, anti-oxidant, anti-tumor and immunomodulation. Their low-toxicity, non-immunogenicity, and biodegradability make them an excellent material in biomedicine. Alginate oligosaccharides can be chemically or biochemically modified to enhance their biological activity and potential in pharmaceutical applications. This paper provides a brief overview on alginate oligosaccharides characteristics, modification patterns and highlights their vital health promoting properties.
Collapse
Affiliation(s)
- Mingpeng Wang
- College of Life Science, Qufu Normal University, Qufu 273100, China
| | - Lei Chen
- College of Life Science, Qufu Normal University, Qufu 273100, China.
| | - Zhaojie Zhang
- Department of Zoology and Physiology, University of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|
41
|
Shteinberg M, Haq IJ, Polineni D, Davies JC. Cystic fibrosis. Lancet 2021; 397:2195-2211. [PMID: 34090606 DOI: 10.1016/s0140-6736(20)32542-3] [Citation(s) in RCA: 327] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/03/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis is a monogenic disease considered to affect at least 100 000 people worldwide. Mutations in CFTR, the gene encoding the epithelial ion channel that normally transports chloride and bicarbonate, lead to impaired mucus hydration and clearance. Classical cystic fibrosis is thus characterised by chronic pulmonary infection and inflammation, pancreatic exocrine insufficiency, male infertility, and might include several comorbidities such as cystic fibrosis-related diabetes or cystic fibrosis liver disease. This autosomal recessive disease is diagnosed in many regions following newborn screening, whereas in other regions, diagnosis is based on a group of recognised multiorgan clinical manifestations, raised sweat chloride concentrations, or CFTR mutations. Disease that is less easily diagnosed, and in some cases affecting only one organ, can be seen in the context of gene variants leading to residual protein function. Management strategies, including augmenting mucociliary clearance and aggressively treating infections, have gradually improved life expectancy for people with cystic fibrosis. However, restoration of CFTR function via new small molecule modulator drugs is transforming the disease for many patients. Clinical trial pipelines are actively exploring many other approaches, which will be increasingly needed as survival improves and as the population of adults with cystic fibrosis increases. Here, we present the current understanding of CFTR mutations, protein function, and disease pathophysiology, consider strengths and limitations of current management strategies, and look to the future of multidisciplinary care for those with cystic fibrosis.
Collapse
Affiliation(s)
- Michal Shteinberg
- Pulmonology Institute and CF Center, Carmel Medical Center, Haifa, Israel; Rappaport Faculty of Medicine, The Technion-Israel Institute of Technology, Haifa, Israel
| | - Iram J Haq
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Jane C Davies
- National Heart and Lung Institute, Imperial College London, London, UK; Royal Brompton and Harefield, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| |
Collapse
|
42
|
Asadpoor M, Varasteh S, Pieters RJ, Folkerts G, Braber S. Differential effects of oligosaccharides on the effectiveness of ampicillin against Escherichia coli in vitro. PHARMANUTRITION 2021. [DOI: 10.1016/j.phanu.2021.100264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Park RM, Nguyen NHT, Lee SM, Kim YH, Min J. Alginate oligosaccharides can maintain activities of lysosomes under low pH condition. Sci Rep 2021; 11:11504. [PMID: 34075195 PMCID: PMC8169924 DOI: 10.1038/s41598-021-91175-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/18/2021] [Indexed: 12/24/2022] Open
Abstract
The objective of this study was to report that lysosome extracted from egg white could be used as a drug through oral administration for treating diseases by using pH sensitive alginate oligosaccharides. Lysosome-alginate oligosaccharides composite were formulated for oral administration of lysosomes. The dissolution test confirmed the availability of the oral dosage form. When lysosome were used as an independent drug, the activity of protein was lost due to influence of low pH. Its antibacterial activity was also remarkably reduced. However, when lysosome-alginate oligosaccharides composite form was used, antimicrobial activity of lysozyme was maintained. At low pH, a gel-like matrix was formed by alginate oligosaccharides to protect the lysosome. When the pH was increased, alginate oligosaccharides were dissolved and the lysosome was released. SDS–polyacrylamide gel electrophoresis analysis of released lysosomes revealed that alginate oligosaccharide could effectively protect the lysosome from degradation or hydrolysis under acidic conditions for at least 2 h. The results of this study are important for application of lysosomes as therapeutic agents, and also it was confirmed that alginate oligosaccharides have potential as direct delivery system for the oral application of protein derived therapies.
Collapse
Affiliation(s)
- Ra-Mi Park
- Graduate School of Semiconductors and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| | - Ngoc-Han Thi Nguyen
- Department of Bioprocess Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| | - Su-Min Lee
- Graduate School of Semiconductors and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, Chungdae-Ro, Seowon-Gu, Cheongju, 28644, South Korea.
| | - Jiho Min
- Graduate School of Semiconductors and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea. .,Department of Bioprocess Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea.
| |
Collapse
|
44
|
Chiron E, Varache M, Stokniene J, Thomas DW, Ferguson EL. A physicochemical assessment of the thermal stability of dextrin-colistin conjugates. Sci Rep 2021; 11:10600. [PMID: 34011987 PMCID: PMC8134461 DOI: 10.1038/s41598-021-89946-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 11/25/2022] Open
Abstract
Attachment of polysaccharide carriers is increasingly being used to achieve precision delivery and improved effectiveness of protein and peptide drugs. Although it is clear that their clinical effectiveness relies on the purity and integrity of the conjugate in storage, as well as following administration, instability of polysaccharide-based conjugates can reduce the protective efficacy of the polymer, which may adversely affect the bioactive's potency. As a model, these studies used dextrin-colistin conjugates, with varying degrees of polymer modification (1, 2.5 and 7.5 mol% succinoylation) to assess the effect of storage temperature (- 20, 4, 21 and 37 °C) and duration (up to 12 months) on saccharide and colistin release and antimicrobial activity. Estimation of the proportion of saccharide release (by comparison of area under the curve from size exclusion chromatograms) was more pronounced at higher temperatures (up to 3 and 35% at - 20 °C and 37 °C, respectively after 12 months), however, repeated freeze-thaw did not produce any measurable release of saccharides, while addition of amylase (20, 100, 500 IU/L) caused rapid release of saccharides (> 70% total within 24 h). At all temperatures, conjugates containing the lowest degree of succinoylation released the highest proportion of free colistin, which increased with storage temperature, however no trend in saccharide release was observed. Despite the clear physical effects of prolonged storage, antimicrobial activity of all samples was only altered after storage at 37 °C for 12 months (> threefold decreased activity). These results demonstrate significant release of saccharides from dextrin-colistin conjugates during prolonged storage in buffered solution, especially at elevated temperature, which, in most cases, did not affect antimicrobial activity. These findings provide vital information about the structure-activity relationship of dextrin-colistin conjugates, prior to full-scale commercial development, which can subsequently be applied to other polysaccharide-protein and -peptide conjugates.
Collapse
Affiliation(s)
- Emilie Chiron
- Advanced Therapies Group, Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff, CF14 4XY, UK
| | - Mathieu Varache
- Advanced Therapies Group, Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff, CF14 4XY, UK
- CNRS, Ingénierie Des Matériaux Polymères, IMP UMR CNRS 5223, Université de Lyon, Université Claude Bernard Lyon 1, 15 bd A. Latarjet, 69622, Villeurbanne, France
| | - Joana Stokniene
- Advanced Therapies Group, Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff, CF14 4XY, UK
| | - David W Thomas
- Advanced Therapies Group, Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff, CF14 4XY, UK
| | - Elaine L Ferguson
- Advanced Therapies Group, Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff, CF14 4XY, UK.
| |
Collapse
|
45
|
Mrudulakumari Vasudevan U, Lee OK, Lee EY. Alginate derived functional oligosaccharides: Recent developments, barriers, and future outlooks. Carbohydr Polym 2021; 267:118158. [PMID: 34119132 DOI: 10.1016/j.carbpol.2021.118158] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023]
Abstract
Alginate is a biopolymer used extensively in the food, pharmaceutical, and chemical industries. Alginate oligosaccharides (AOS) derived from alginate exhibit superior biological activities and therapeutic potential. Alginate lyases with characteristic substrate specificity can facilitate the production of a broad array of AOS with precise structure and functionality. By adopting innovative analytical tools in conjunction with focused clinical studies, the structure-bioactivity relationship of a number of AOS has been brought to light. This review covers fundamental aspects and recent developments in AOS research. Enzymatic and microbial processes involved in AOS production from brown algae and sequential steps involved in AOS structure elucidation are outlined. Biological mechanisms underlying the health benefits of AOS and their potential industrial and therapeutic applications are elaborated. Withal, various challenges in AOS research are traced out, and future directions, specifically on recombinant systems for AOS preparation, are delineated to further widen the horizon of these exceptional oligosaccharides.
Collapse
Affiliation(s)
- Ushasree Mrudulakumari Vasudevan
- Department of Chemical Engineering (Integrated Engineering), Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Ok Kyung Lee
- Department of Chemical Engineering (Integrated Engineering), Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Eun Yeol Lee
- Department of Chemical Engineering (Integrated Engineering), Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| |
Collapse
|
46
|
Abstract
Antimicrobial resistance is a serious medical threat, particularly given the decreasing rate of discovery of new treatments. Although attempts to find new treatments continue, it has become clear that merely discovering new antimicrobials, even if they are new classes, will be insufficient. It is essential that new strategies be aggressively pursued. Toward that end, the search for treatments that can mitigate bacterial virulence and tilt the balance of host-pathogen interactions in favor of the host has become increasingly popular. In this review, we will discuss recent progress in this field, with a special focus on synthetic small molecule antivirulents that have been identified from high-throughput screens and on treatments that are effective against the opportunistic human pathogen Pseudomonas aeruginosa.
Collapse
|
47
|
Stuart-Walker W, Mahon CS. Glycomacromolecules: Addressing challenges in drug delivery and therapeutic development. Adv Drug Deliv Rev 2021; 171:77-93. [PMID: 33539854 DOI: 10.1016/j.addr.2021.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/15/2021] [Accepted: 01/23/2021] [Indexed: 12/18/2022]
Abstract
Carbohydrate-based materials offer exciting opportunities for drug delivery. They present readily available, biocompatible components for the construction of macromolecular systems which can be loaded with cargo, and can enable targeting of a payload to particular cell types through carbohydrate recognition events established in biological systems. These systems can additionally be engineered to respond to environmental stimuli, enabling triggered release of payload, to encompass multiple modes of therapeutic action, or to simultaneously fulfil a secondary function such as enabling imaging of target tissue. Here, we will explore the use of glycomacromolecules to deliver therapeutic benefits to address key health challenges, and suggest future directions for development of next-generation systems.
Collapse
|
48
|
Asadpoor M, Ithakisiou GN, Henricks PAJ, Pieters R, Folkerts G, Braber S. Non-Digestible Oligosaccharides and Short Chain Fatty Acids as Therapeutic Targets against Enterotoxin-Producing Bacteria and Their Toxins. Toxins (Basel) 2021; 13:175. [PMID: 33668708 PMCID: PMC7996226 DOI: 10.3390/toxins13030175] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Enterotoxin-producing bacteria (EPB) have developed multiple mechanisms to disrupt gut homeostasis, and provoke various pathologies. A major part of bacterial cytotoxicity is attributed to the secretion of virulence factors, including enterotoxins. Depending on their structure and mode of action, enterotoxins intrude the intestinal epithelium causing long-term consequences such as hemorrhagic colitis. Multiple non-digestible oligosaccharides (NDOs), and short chain fatty acids (SCFA), as their metabolites produced by the gut microbiota, interact with enteropathogens and their toxins, which may result in the inhibition of the bacterial pathogenicity. NDOs characterized by diverse structural characteristics, block the pathogenicity of EPB either directly, by inhibiting bacterial adherence and growth, or biofilm formation or indirectly, by promoting gut microbiota. Apart from these abilities, NDOs and SCFA can interact with enterotoxins and reduce their cytotoxicity. These anti-virulent effects mostly rely on their ability to mimic the structure of toxin receptors and thus inhibiting toxin adherence to host cells. This review focuses on the strategies of EPB and related enterotoxins to impair host cell immunity, discusses the anti-pathogenic properties of NDOs and SCFA on EPB functions and provides insight into the potential use of NDOs and SCFA as effective agents to fight against enterotoxins.
Collapse
Affiliation(s)
- Mostafa Asadpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Georgia-Nefeli Ithakisiou
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Paul A. J. Henricks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Roland Pieters
- Division of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands;
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| |
Collapse
|
49
|
Martin I, Waters V, Grasemann H. Approaches to Targeting Bacterial Biofilms in Cystic Fibrosis Airways. Int J Mol Sci 2021; 22:ijms22042155. [PMID: 33671516 PMCID: PMC7926955 DOI: 10.3390/ijms22042155] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
The treatment of lung infection in the context of cystic fibrosis (CF) is limited by a biofilm mode of growth of pathogenic organisms. When compared to planktonically grown bacteria, bacterial biofilms can survive extremely high levels of antimicrobials. Within the lung, bacterial biofilms are aggregates of microorganisms suspended in a matrix of self-secreted proteins within the sputum. These structures offer both physical protection from antibiotics as well as a heterogeneous population of metabolically and phenotypically distinct bacteria. The bacteria themselves and the components of the extracellular matrix, in addition to the signaling pathways that direct their behaviour, are all potential targets for therapeutic intervention discussed in this review. This review touches on the successes and failures of current anti-biofilm strategies, before looking at emerging therapies and the mechanisms by which it is hoped they will overcome current limitations.
Collapse
Affiliation(s)
- Isaac Martin
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada;
- Correspondence:
| | - Valerie Waters
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada;
- Department of Paediatrics and Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Hartmut Grasemann
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada;
- Department of Paediatrics and Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
50
|
Ci F, Jiang H, Zhang Z, Mao X. Properties and potential applications of mannuronan C5-epimerase: A biotechnological tool for modifying alginate. Int J Biol Macromol 2021; 168:663-675. [PMID: 33220370 DOI: 10.1016/j.ijbiomac.2020.11.123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 11/23/2022]
Abstract
Given the excellent characteristics of alginate, it is an industrially important polysaccharide. Mannuronan C5-epimerase (MC5E) is an alginate-modifying enzyme that catalyzes the conversion of β-D-mannuronate (M) to its C5 epimer α-L-guluronate (G) in alginate. Both the biological activities and physical properties of alginate are determined by M/G ratios and distribution patterns. Therefore, MC5E is regarded as a biotechnological tool for modifying and processing alginate. Various MC5Es derived from brown algae, Pseudomonas and Azotobacter have been isolated and characterized. With the rapid development of structural biology, the crystal structures and catalytic mechanisms of several MC5Es have been elucidated. It is necessary to comprehensively understand the research status of this alginate-modifying enzyme. In this review, the properties and potential applications of MC5Es isolated from different kinds of organisms are summarized and reviewed. Moreover, future research directions of MC5Es as well as strategies to enhance their properties are elucidated, highlighted, and prospected.
Collapse
Affiliation(s)
- Fangfang Ci
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Hong Jiang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| | - Zhaohui Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Xiangzhao Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|