Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Enzyme Microb Technol ; 177: 110426, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38503081

RESUMEN

Eukaryotic sialyltransferases play key roles in many physiological and pathological events. The expression of active human recombinant sialyltransferases in bacteria is still challenging. In the current study, the genes encoding human N-acetylgalactosaminide α2,6-sialyltransferase V (hST6GalNAc V) and N-acetylgalactosaminide α2,6-sialyltransferase VI (hST6GalNAc VI) lacking the N-terminal transmembrane domains were cloned into the expression vectors, pET-32a and pET-22b, respectively. Soluble and active forms of recombinant hST6GalNAc V and hST6GalNAc VI when coexpressed with the chaperone plasmid pGro7 were successfully achieved in Escherichia coli. Further, lactose (Lac), Lacto-N-triose II (LNT II), lacto-N-tetraose (LNT), and sialyllacto-N-tetraose a (LSTa) were used as acceptor substrates to investigate their activities and substrate specificities. Unexpectedly, both can transfer sialic acid onto all those substrates. Compared with hST6GalNAc V expressed in the mammalian cells, the recombinant two α2,6-sialyltransferases in bacteria displayed flexible substrate specificities and lower enzymatic efficiency. In addition, an important human milk oligosaccharide disialyllacto-N-tetraose (DSLNT) can be synthesized by both human α2,6-sialyltransferases expressed in E. coli using LSTa as an acceptor substrate. To the best of our knowledge, these two active human α2,6-sialyltransferases enzymes were expressed in bacteria for the first time. They showed a high potential to be applied in biotechnology and investigating the molecular mechanisms of biological and pathological interactions related to sialylated glycoconjugates.


Asunto(s)
Escherichia coli , Proteínas Recombinantes , Sialiltransferasas , Humanos , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Lactosa/metabolismo , Oligosacáridos/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Especificidad por Sustrato
2.
Carbohydr Polym ; 333: 121999, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38494241

RESUMEN

Chitosan and chitooligosaccharide (COS) are renowned for their potent antimicrobial prowess, yet the precise antimicrobial efficacy of COS remains elusive due to scant structural information about the utilized saccharides. This study delves into the antimicrobial potential of COS, spotlighting a distinct hetero-chitooligosaccharide dubbed DACOS. In contrast to other COS, DACOS remarkably fosters the growth of Candida tropicalis planktonic cells and fungal biofilms. Employing gradient alcohol precipitation, DACOS was fractionated, unveiling diverse structural characteristics and differential impacts on C. tropicalis. Notably, in a murine model of systemic candidiasis, DACOS, particularly its 70 % alcohol precipitates, manifests a promotive effect on Candida infection. This research unveils a new pathway for exploring the intricate nexus between the structural attributes of chitosan oligosaccharides and their physiological repercussions, underscoring the imperative of crafting chitosan and COS with meticulously defined structural configurations.


Asunto(s)
Antiinfecciosos , Quitosano , Oligosacáridos , Animales , Ratones , Candida tropicalis , Quitosano/farmacología , Quitosano/química , Antifúngicos/farmacología , Biopelículas
3.
Sheng Wu Gong Cheng Xue Bao ; 39(10): 4135-4149, 2023 Oct 25.
Artículo en Chino | MEDLINE | ID: mdl-37877396

RESUMEN

The biofilms formed by pathogenic microorganisms seriously threaten human health and significantly enhance drug resistance, which urgently call for developing drugs specifically targeting on biofilms. Chitooligosaccharides extracted from shrimp and crab shells are natural alkaline oligosaccharides with excellent antibacterial effects. Nevertheless, their inhibition efficacy on biofilms still needs to be improved. Spirulina (SP) is a microalga with negatively charged surface, and its spiral structure facilitates colonization in the depth of the biofilm. Therefore, the complex of Spirulina and chitooligosaccharides may play a synergistic role in killing pathogens in the depth of biofilm. This research first screened chitooligosaccharides with significant bactericidal effects. Subsequently, Spirulina@Chitooligosaccharides (SP@COS complex was prepared by combining chitooligosaccharides with Spirulina through electrostatic adsorption. The binding of the complex was characterized by zeta potential, z-average size, and fluorescence labeling. Ultraviolet-visible spectroscopy (UV-Vis) showed the encapsulation efficiency and the drug loading efficiency reached up to 90% and 16%, respectively. The prepared SP@COS2 exhibited a profound synergistic inhibition effect on bacterial and fungal biofilms, which was mainly achieved by destroying the cell structure of the biofilm. These results demonstrate the potential of Spirulina-chitooligosaccharides complex as a biofilm inhibitor and provide a new idea for addressing the harm of pathogenic microorganisms.


Asunto(s)
Quitosano , Spirulina , Humanos , Antibacterianos/farmacología , Antibacterianos/química , Quitosano/farmacología , Biopelículas , Quitina/farmacología
4.
Int J Biol Macromol ; 253(Pt 1): 126627, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37660864

RESUMEN

Glycocalyx dysfunction is believed as the first step in diabetic vascular disease. However, few studies have systematically investigated the influence of HG on the glycocalyx as a whole and its major constituent glycans towards one type of cell. Furthermore, most studies utilized traditional two-dimensional (2D) cultures in vitro, which can't provide the necessary fluid environment for glycocalyx. Here, we utilized vascular glycocalyx on chips to evaluate the changes of glycocalyx and its constituent glycans in HG induced HUVECs. Fluorescence microscopy showed up-regulation of hyaluronan (HA) but down-regulation of heparan sulfate (HS). By analyzing the metabolic enzymes of both glycans, a decrease in the ratio of synthetic/degradative enzymes for HA and an increase in that for HS were demonstrated. Two substrates (UDP-GlcNAc, UDP-GlcA) for the synthesis of both glycans were increased according to omics analysis. Since they were firstly pumped into Golgi apparatus to synthesize HS, less substrates may be left for HA synthesis. Furthermore, the differential changes of HA and HS were confirmed in vessel slides from db/db mice. This study would deepen our understanding of impact of HG on glycocalyx formation and diabetic vascular disease.


Asunto(s)
Angiopatías Diabéticas , Ácido Hialurónico , Ratones , Animales , Ácido Hialurónico/metabolismo , Heparitina Sulfato/metabolismo , Glucosa , Uridina Difosfato
5.
Front Chem ; 10: 871509, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572116

RESUMEN

The pandemic caused by SARS-CoV-2 is the most widely spread disease in the 21st century. Due to the continuous emergence of variants across the world, it is necessary to expand our understanding of host-virus interactions and explore new agents against SARS-CoV-2. In this study, it was found exopolysaccharides (EPSs) from halophilic archaeon Haloarcula hispanica ATCC33960 can bind to the spike protein of SARS-CoV-2 with the binding constant KD of 2.23 nM, block the binding of spike protein to Vero E6 and bronchial epithelial BEAS-2B cells, and inhibit pseudovirus infection. However, EPSs from the gene deletion mutant △HAH_1206 almost completely lost the antiviral activity against SARS-CoV-2. A significant reduction of glucuronic acid (GlcA) and the sulfation level in EPSs of △HAH_1206 was clearly observed. Our results indicated that sulfated GlcA in EPSs is possible for a main structural unit in their inhibition of binding of SARS-CoV-2 to host cells, which would provide a novel antiviral mechanism and a guide for designing new agents against SARS-CoV-2.

6.
Front Cell Dev Biol ; 10: 877892, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35557948

RESUMEN

Past studies on the protective effects of chitosan oligosaccharides (COS) on inflammatory bowel disease (IBD) commonly rely on animal models, because traditional cell culture systems couldn't faithfully mimic human intestinal physiology. Here a novel human gut-on-a-chip microsystem was established to further explore the regulatory effects of COS on the occurrence and development of human enteritis. By constructing an intestinal injury model caused by dextran sodium sulfate (DSS) on the chip, this study proved that COS can reduce intestinal epithelial injury by promoting the expression of the mucous layer for the first time. By establishing an inflammatory bowel disease model on the chip caused by E. coli 11775, this study demonstrated that COS can protect the intestinal epithelial barrier and vascular endothelial barrier by inhibiting the adhesion and invasion of E. coli 11775 for the first time. In addition, similar to the results in vivo, COS can decrease the inflammatory response by reducing the expression of toll-like receptor 4 protein and reducing the nuclear DNA binding rate of nuclear factor kappa-B protein on this chip. In summary, COS can be used as a potential drug to treat human IBD and the human gut-on-a-chip would be used as a platform for quick screening drugs to treat human IBD in future.

7.
Carbohydr Polym ; 285: 119253, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35287867

RESUMEN

The fungal cell wall is an ideal target for the design of antifungal drugs. In this study we used an analog of cell wall polymer, a highly deacetylated high molecular-weight chitosan oligosaccharide (HCOS), to test its effect against pathogenic Candida strains. Results showed that HCOS was successfully incorporated into the dynamic cell wall organization process and exhibited an apparent antifungal activity against both plankton and mature fungal biofilm, by impairing the cell wall integrity. Unexpectedly, mechanistic studies suggested that HCOS exerts its activity by interfering with family members of PHR ß-(1,3)-glucanosyl transferases and affecting the connection and assembly of cell wall polysaccharides. Furthermore, HCOS showed great synergistic activity with different fungicides against Candida cells, especially those in biofilm. These findings indicated HCOS has a great potential as an antifungal drug or drug synergist and proposed a novel antifungal strategy with structure-specific oligosaccharides mimicking cell wall polysaccharide fragments.


Asunto(s)
Antifúngicos , Quitosano , Antifúngicos/farmacología , Biopelículas , Candida albicans , Pared Celular , Quitosano/farmacología , Pruebas de Sensibilidad Microbiana , Peso Molecular , Oligosacáridos/farmacología
8.
J Biomater Sci Polym Ed ; 33(4): 517-531, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34704529

RESUMEN

Osteoarthritis (OA) is a degenerative joint disease, which can lead to joint pain, stiffness, deformity and dysfunction, that seriously affects the quality of life in patients. At present, the treatments of OA mainly include early pharmacological treatment and late joint replacement. However, current pharmacological treatment has limited efficacy and undesired side effects. Chitosan oligosaccharide (COS) is a kind of nontoxic and biodegradable oligo-saccharide, which is composed of 2-20 glucosamine or N-acetylglucosamine linked by ß-1,4 glycosidic bond. Studies have shown that COS has significant biological properties like antimicrobial, anti-inflammatory, antioxidant, and anti-tumor, as well as immunoregulation ability. However, the effects of COS on OA have not been clarified. In this study, we explored the protective effects of COS with different degrees of deacetylation on chondrocytes stimulated by interleukin 1ß (IL-1ß) in vitro. The results showed that IL-1ß inhibited cell proliferation and promoted cell apoptosis. Besides that, IL-1ß increased the expression of the major chondro-degrading genes MMP13 and ADAMTS-5, while decreased the expression of COL2A and ACAN. COS with different degrees of deacetylation (HDACOS, MDACOS, LDACOS) had different effects on IL-1ß induced inflammation. LDACOS had the most obvious anti-inflammatory effects to inhibit the expression of MMP13 and ADAMTS-5 while promoted the expression of COL2A and ACAN. In addition, we found that the expression of autophagy-related gene Beclin-1 was up-regulated, and the ratio of LC3-II/LC3-I was increased in the LDACOS group. Furthermore, transmission electron microscopy (TEM) analysis showed that the number of intracellular autophagosomes increased significantly with the treatment of LDACOS. Based on our research, we suggested that LDACOS could inhibit chondrocytes inflammation and promote cell autophagy, and might be a protective drug for the treatment of OA.


Asunto(s)
Quitosano , Osteoartritis , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Autofagia , Quitosano/metabolismo , Quitosano/farmacología , Condrocitos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucina-1beta/uso terapéutico , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Oligosacáridos/farmacología , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Osteoartritis/patología , Calidad de Vida
9.
Front Pharmacol ; 12: 694107, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34149435

RESUMEN

Glucosamine (GlcN) is used as a supplement for arthritis and joint pain and has been proved to have effects on inflammation, cancer, and cardiovascular diseases. However, there are limited studies on the regulatory mechanism of GlcN against glucose and lipid metabolism disorder. In this study, we treated high-fat diet (HFD)-induced diabetic mice with GlcN (1 mg/ml, in drinking water) for five months. The results show that GlcN significantly reduced the fasting blood glucose of HFD-fed mice and improved glucose tolerance. The feces of intestinal contents in mice were analyzed using 16s rDNA sequencing. It was indicated that GlcN reversed the imbalanced gut microbiota in HFD-fed mice. Based on the PICRUSt assay, the signaling pathways of glucolipid metabolism and biosynthesis were changed in mice with HFD feeding. By quantitative real-time PCR (qPCR) and hematoxylin and eosin (H&E) staining, it was demonstrated that GlcN not only inhibited the inflammatory responses of colon and white adipose tissues, but also improved the intestinal barrier damage of HFD-fed mice. Finally, the correlation analysis suggests the most significantly changed intestinal bacteria were positively or negatively related to the occurrence of inflammation in the colon and fat tissues of HFD-fed mice. In summary, our studies provide a theoretical basis for the potential application of GlcN to glucolipid metabolism disorder through the regulation of gut microbiota.

10.
Mar Drugs ; 19(6)2021 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-34073769

RESUMEN

It is known that bioactivities of chitooligosaccharide (COS) are closely related to the degree of polymerization (DP); therefore, it is essential to prepare COS with controllable DP, such as chitobiose showing high antioxidant and antihyperlipidemia activities. In this study, BLAST, sequence alignment and phylogenetic analysis of characterized glycoside hydrolase (GH) 46 endo-chitosanases revealed that a chitosanase Sn1-CSN from Streptomyces niveus was different from others. Sn1-CSN was overexpressed in E. coli, purified and characterized in detail. It showed the highest activity at pH 6.0 and exhibited superior stability between pH 4.0 and pH 11.0. Sn1-CSN displayed the highest activity at 50 °C and was fairly stable at ≤45 °C. Its apparent kinetic parameters against chitosan (DDA: degree of deacetylation, >94%) were determined, with Km and kcat values of 1.8 mg/mL and 88.3 s-1, respectively. Cu2+ enhanced the activity of Sn1-CSN by 54.2%, whereas Fe3+ inhibited activity by 15.1%. Hydrolysis products of chitosan (DDA > 94%) by Sn1-CSN were mainly composed of chitobiose (87.3%), whereas partially acetylated chitosan with DDA 69% was mainly converted into partially acetylated COS with DP 2-13. This endo-chitosanase has great potential to be used for the preparation of chitobiose and partially acetylated COS with different DPs.


Asunto(s)
Glicósido Hidrolasas , Streptomyces/enzimología , Secuencia de Aminoácidos , Quitosano/química , Quitosano/metabolismo , Disacáridos/metabolismo , Escherichia coli/genética , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Concentración de Iones de Hidrógeno , Hidrólisis , Metales/química , Simulación del Acoplamiento Molecular , Filogenia , Temperatura
11.
Food Chem ; 353: 129460, 2021 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-33725543

RESUMEN

Endo-fucoidanases are important in structural analysis of fucoidans and preparation of fuco-oligosaccharides. However their enzymological properties and analysis of degradation products are scarcely investigated. Truncated endo-α (1 â†’ 3)-fucoidanase Fda1 (tFda1B from Alteromonas sp. was overexpressed and characterized, showing highest activity at pH 7.0, 35 °C, and 1.0 M NaCl. Its Km and kcat were 3.88 ± 0.81 mg/mL and 0.82 ± 0.17 min-1. Fe3+ and Mn2+ enhanced activity by 100% and 19.5% respectively. Co2+ and Cu2+ completely inactivated tFda1B, whereas Ni2+, Mg2+, Zn2+, Pb2+, Ca2+, Ba2+ and Li+ decreased activity by 58.8%, 56.0%, 50.6%, 47.7%, 28.9%, 15.6% and 37.5%, respectively. Catalytic residues were identified through structure and sequence alignment, and confirmed by mutagenesis. Degradation products of Kjellmaniella crassifolia fucoidan by tFda1B were characterized by LC-ESI-MS/MS, confirming tFda1B belongs to endo-(1 â†’ 3)-fucoidanases, and backbone of K. crassifolia fucoidan is 1 â†’ 3 fucoside linkage. This endo-α (1 â†’ 3)-fucoidanase would be useful for elucidating fucoidan structures, and be used as a food enzyme.


Asunto(s)
Alteromonas/enzimología , Hidrolasas/química , Hidrolasas/metabolismo , Polisacáridos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Estabilidad de Enzimas , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Concentración de Iones de Hidrógeno , Hidrolasas/genética , Mutagénesis Sitio-Dirigida , Oligosacáridos/química , Phaeophyceae/química , Phaeophyceae/metabolismo , Filogenia , Polisacáridos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Especificidad por Sustrato , Espectrometría de Masas en Tándem
12.
Molecules ; 25(24)2020 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-33371321

RESUMEN

The formation of bacterial biofilms has increased the resistance of bacteria to various environmental factors and is tightly associated with many persistent and chronic bacterial infections. Herein we design a strategy conjugating florfenicol, an antibiotic commonly used in the treatment of streptococcus, with the antimicrobial biomaterial, chitosan oligosaccharides. The results demonstrated that the florfenicol-COS conjugate (F-COS) efficiently eradicated the mature Streptococcus hyovaginalis biofilm, apparently inhibiting drug resistance to florfenicol. A quantity of 250 µg/mL F-COS showed effective inhibitory activity against planktonic cells and biofilm of the bacteria, and a 4-fold improvement of the F-COS compared to unmodified florfenicol was observed. Furthermore, the conjugate showed a broad-spectrum activity against both Gram-positive and Gram-negative bacteria. It suggested that F-COS might have a potential for application in the treatment of biofilm-related infections.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Quitosano/química , Farmacorresistencia Bacteriana/efectos de los fármacos , Oligosacáridos/química , Oligosacáridos/farmacología , Infecciones Bacterianas/tratamiento farmacológico , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Plancton/efectos de los fármacos , Streptococcus/efectos de los fármacos , Tianfenicol/análogos & derivados , Tianfenicol/química , Tianfenicol/farmacología
13.
Mar Drugs ; 18(10)2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32992800

RESUMEN

It is proven that ß-amyloid (Aß) aggregates containing cross-ß-sheet structures led to oxidative stress, neuroinflammation, and neuronal loss via multiple pathways. Therefore, reduction of Aß neurotoxicity via inhibiting aggregation of Aß or dissociating toxic Aß aggregates into nontoxic forms might be effective therapeutic methods for Alzheimer's disease (AD) treatment. This study was designed to explore interference of chitosan oligosaccharides (COS) on ß-(1-42)-amyloid protein (Aß42) aggregation and Aß42-induced cytotoxicity. Here it was demonstrated that COS showed good blood-brain barrier (BBB) penetration ability in vitro and in vivo. The experimental results showed that COS efficiently interfered with Aß42 aggregation in dose- and degree of polymerization (DP)-dependent manners, and COS monomer with DP6 showed the best effect on preventing conformational transition into ß-sheet-rich structures. Based on the binding affinity analysis by microscale thermophoresis (MST), it was confirmed that COS could directly bind with Aß42 in a DP-dependent manner. Our findings demonstrated that different performance of COS monomers with different DPs against Aß42 assembly was, to some extent, attributable to their different binding capacities with Aß42. As a result, COS significantly ameliorated Aß42-induced cytotoxicity. Taken together, our studies would point towards a potential role of COS in treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/metabolismo , Quitosano/química , Oligosacáridos/administración & dosificación , Fragmentos de Péptidos/metabolismo , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oligosacáridos/farmacocinética , Oligosacáridos/farmacología , Estrés Oxidativo/efectos de los fármacos , Polimerizacion , Distribución Tisular
14.
Int J Pharm ; 587: 119669, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32702454

RESUMEN

Organ-on-a-chip as a new technology distinguishes it from animal and cell models at least in three aspects: (1) it responds to drugs' efficacy or toxicity more really by mimicking the human body's fluid microenvironment; (2) it can be used for high throughput screening a large number of compounds; (3) it has physiological accuracy. It is well known that ginsenosides compound K (CK) as a carbohydrate drug has numerous biological activities and physiological functions. However, pharmacokinetic studies of carbohydrate-based CK haven't been performed on organ chips. Here, we established and evaluated the function of single-organ chips and multi-organ chips based on intestinal, vascular, liver, and kidney chips. Each single-organ-on-a-chip performed itself well. Based on organ-on-chips, absorption, metabolism and toxicity of CK were successfully investigated. The pharmacokinetic results of CK provided by chip were consistent with previous reports, demonstrating the reliability of the organ-on-a-chip platform and its potential for use in pharmacokinetic studies of carbohydrate-drugs. As far as we know, this study would be the first report on the pharmacological investigation of carbohydrate drugs on organ-on-a-chip, which provides a theoretical basis for carbohydrate-based drug discovery.


Asunto(s)
Ginsenósidos , Animales , Ginsenósidos/toxicidad , Humanos , Riñón , Dispositivos Laboratorio en un Chip , Reproducibilidad de los Resultados
15.
Carbohydr Polym ; 220: 60-70, 2019 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-31196551

RESUMEN

Chitosan oligosaccharides (COS) are the degraded products of chitin or chitosan prepared by chemical or enzymatic hydrolysis. As compared to chitosan, COS not only exhibit some specific physicochemical properties such as excellent water solubility, biodegradability and biocompatibility, but also have a variety of functionally biological activities including anti-inflammation, anti-bacteria, immunomodulation, neuroprotection and so on. This review aims to summarize the preparation and structural characterization methods of COS, and will discuss the application of COS or their derivatives to human health, animal husbandry and agricultural production. COS have been demonstrated to prevent the occurrence of human health-related diseases, enhance the resistance to diseases of livestock and poultry, and improve the growth and quality of crops in plant cultivation. Overall, COS have presented a broad developmental potential and application prospect in the healthy field that deserves further exploration.


Asunto(s)
Quitosano , Crianza de Animales Domésticos , Animales , Antiinfecciosos/uso terapéutico , Antiinflamatorios/uso terapéutico , Fármacos Antiobesidad/uso terapéutico , Antineoplásicos/uso terapéutico , Antioxidantes/uso terapéutico , Quitosano/química , Quitosano/aislamiento & purificación , Quitosano/farmacología , Productos Agrícolas/crecimiento & desarrollo , Productos Agrícolas/metabolismo , Humanos , Ganado/crecimiento & desarrollo , Ganado/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Enfermedades de las Plantas/prevención & control
16.
J Diabetes ; 11(1): 32-45, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29845722

RESUMEN

BACKGROUND: N-Acetylcysteine (NAC), an antioxidative reagent for clinical diseases, shows potential in the treatment of diabetes and other metabolic diseases. However, it is unknown how NAC modulates the gut microbiota of mice with metabolic syndrome. The aim of the present study was to demonstrate the preventive effect of NAC on intestinal dysbiosis and glucose metabolic disorder. METHODS: Mice (C57BL/6J strain) were fed either a normal chow diet (NCD), NCD plus NAC, a high-fat diet (HFD), or HFD plus NAC for 5 months, after which glucose levels, circulating endotoxins and key metabolism-related proteins were determined. Fecal samples were analyzed by 16S rRNA sequencing. A novel analysis was performed to predict functional changes in gut microbiota. In addition, Spearman's correlation analysis was performed between metabolic biomarkers and bacterial abundance. RESULTS: Treatment with NAC significantly reversed the glucose intolerance, fasting glucose concentrations, and gains in body weight and plasma endotoxin in HFD-fed mice. Further, NAC upregulated occludin and mucin glycoprotein levels in the proximal colon of HFD-treated mice. Noticeably, NAC promoted the growth of beneficial bacteria (i.e. Akkermansia, Bifidobacterium, Lactobacillus and Allobaculum) and decreased populations of diabetes-related genera, including Desulfovibrio and Blautia. In addition, NAC may affect the metabolic pathways of intestinal bacteria, including lipopolysaccharide biosynthesis, oxidative stress, and bacterial motility. Finally, the modified gut microbiota was closely associated with the metabolic changes in NAC-treated HFD-fed mice. CONCLUSIONS: N-Acetylcysteine may be a potential drug to prevent glucose metabolic disturbances by reshaping the structure of the gut microbiota.


Asunto(s)
Acetilcisteína/farmacología , Dieta Alta en Grasa/efectos adversos , Disbiosis/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Trastornos del Metabolismo de la Glucosa/prevención & control , Acetilcisteína/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/genética , Glucemia/análisis , Disbiosis/metabolismo , Disbiosis/microbiología , Heces/microbiología , Depuradores de Radicales Libres/administración & dosificación , Depuradores de Radicales Libres/farmacología , Microbioma Gastrointestinal/genética , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/prevención & control , Trastornos del Metabolismo de la Glucosa/etiología , Trastornos del Metabolismo de la Glucosa/microbiología , Hiperglucemia/sangre , Hiperglucemia/prevención & control , Lipopolisacáridos/sangre , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN
17.
Carbohydr Polym ; 206: 403-411, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30553339

RESUMEN

The polysaccharides of Enteromorpha prolifera (PEP) displayed various bioactivities such as anti-viral, anti-inflammatory and immune-regulative effects. However, no studies were performed on the biological effect of Enteromorpha prolifera oligomers (EPO). In this study, we prepared EPO and evaluated their anti-diabetic effect. By enzymatic degradation, EPO were produced from PEP, and the average molecular weight was identified to be 44.1 kDa by Gel Permeation Chromatography (GPC) analysis. The major monosaccharide units of EPO were measured to be rhamnose, glucuronic acid, glucose, xylose and galactose by capillary electrophoresis assay. Based on the in vitro studies, EPO presented potent reducing power and antioxidant effect such as the scavenging of 1, 1-diphenyl-2-picrylhydrazyl (DPPH), superoxide and NO radicals. The in vivo studies show that EPO relieved the symptoms of polydipsia, polyphagia, emaciation and hyperglycemia in streptozotocin (STZ)-induced diabetic mice to a certain extent. Further, by using the quantitative real-time PCR (qPCR) assay and immunofluorescence staining, EPO was proved to promote the insulin secretion by reducing pancreatic inflammation and apoptosis in diabetic mice. In summary, our results indicate that the mitigation of EPO on pancreatic damage might be an effective way to ameliorate the diabetes mellitus.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Depuradores de Radicales Libres/uso terapéutico , Hipoglucemiantes/uso terapéutico , Oligosacáridos/uso terapéutico , Ulva/química , Animales , Apoptosis/efectos de los fármacos , Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Depuradores de Radicales Libres/química , Hidrólisis , Hipoglucemiantes/química , Inflamación/tratamiento farmacológico , Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Oligosacáridos/química , Oxidación-Reducción , Páncreas/efectos de los fármacos , Páncreas/patología , Polisacáridos/química , Estreptozocina
18.
Mar Drugs ; 16(11)2018 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-30463189

RESUMEN

Chitosan oligosaccharides (COS) display various biological activities. In this study, we aimed to explore the preventive effects of COS on glucolipid metabolism disorder using palmitic acid (PA)-induced HepG2 cells and high-fat diet (HFD)-fed C57BL/6J mice as experimental models in vitro and in vivo, respectively. The results showed that COS pretreatment for 12 h significantly ameliorated lipid accumulation in HepG2 cells exposed to PA for 24 h, accompanied by a reversing of the upregulated mRNA expression of proinflammatory cytokines (IL-6, MCP-1, TNF-α) and glucolipid metabolism-related regulators (SCD-1, ACC1, PCK1-α). In addition, COS treatment alleviated glucolipid metabolism disorder in mice fed with HFD for five months, including reduction in body weight and fasting glucose, restoration of intraperitoneal glucose tolerance, and suppression of overexpression of proinflammatory cytokines and glucolipid metabolism-related regulators. Furthermore, our study found that COS pretreatment significantly reversed the downregulation of PPARγ at transcriptional and translational levels in both PA-induced HepG2 cells and liver tissues of HFD-fed mice. In summary, the study suggests that COS can improve glucolipid metabolism disorder by suppressing inflammation and upregulating PPARγ expression. This indicates a novel application of COS in preventing and treating glucolipid metabolism-related diseases.


Asunto(s)
Quitosano/farmacología , Glucolípidos/metabolismo , Síndrome Metabólico/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Oligosacáridos/farmacología , Animales , Quitosano/química , Quitosano/uso terapéutico , Citocinas/inmunología , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Células Hep G2 , Humanos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/inmunología , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/inmunología , Obesidad/metabolismo , Oligosacáridos/química , Oligosacáridos/uso terapéutico , PPAR gamma/metabolismo , Resultado del Tratamiento , Regulación hacia Arriba
19.
Front Microbiol ; 9: 2388, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30356845

RESUMEN

Chitosan oligosaccharides (COS) have shown positive effects on host gut health and influence on intestinal microbial community. However, the bioactivity and mechanism of COS on gut microbiota is still poorly understood. Here, we presented systematic studies of COS on mice fecal/gut microbiota. During in vitro fermentation of COS by mice gut microbiota, total bacterial population significantly decreased after 8-h COS treatment but was returned to the normal level after extended incubation. Consumption of COS and production of SCFAs suggested that COS were utilized by the microbe, although the consumption of chitosan pentasaccharides was obviously slower than others. COS treatments on mice fecal samples caused the decrease of potential pathogenic genera Escherichia/Shigella and the increase of genus Parabacteroides. In vivo animal study indicated that COS reduced population of probiotic genera Lactobacillus, Bifidobacterium and harmful genus Desulfovibrio, and increased abundance of genus Akkermansia. Phylum Proteobacteria was significantly inhibited by COS both in the animal model and in vitro fermentation. Our findings suggested that COS could reform the community structure of gut microbiota. The relationship among COS, gut microbiota and host health deserve further study.

20.
Int J Biol Macromol ; 116: 100-105, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29733934

RESUMEN

Cold-active cellulases have received great attention for both industrial applications and fundamental research because of their high activity at low temperatures and their unique structural characters. In this study, the cold-active endoglucanase CelX from psychrotrophic Pseudoalteromonas sp. DY3 was successfully overexpressed in E. coli, partly purified and characterized in detail. CelX showed the highest activity at pH 5.5, and exhibited moderate activity and superior pH stability over a wide pH range (pH 5.0-pH 9.0). It displayed the highest activity at 45 °C, and kept 34.7% residual activity even at 5 °C. It was stable below 35 °C and lost activity very quickly above 45 °C, which is consistent with its cold adaptability. The apparent kinetic parameters CelX against CMC (carboxymethyl cellulose) were determined, with the Km and kcat values of 6.4 mg/ml and 4.2 s-1 respectively. Mn2+ and Co2+ enhanced the cellulolytic activity of CelX by 28.8% and 20.6% respectively, whereas Pb2+ and Cu2+ inhibited its activity by 14.9% and 6.5% separately. The cold adaptation of CelX is possibly due to the presence of the unusually long linker between the catalytic module and the cellulose-binding domain.


Asunto(s)
Proteínas Bacterianas/genética , Celulasas/genética , Pseudoalteromonas/genética , Carboximetilcelulosa de Sodio/metabolismo , Catálisis , Frío , Escherichia coli/genética , Concentración de Iones de Hidrógeno , Cinética , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...