Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 407
Filter
1.
mSystems ; 9(8): e0052224, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38980058

ABSTRACT

The gut microbiota is essential for providing colonization resistance against pathogens. Dietary sugars markedly shift the composition of the intestinal microbiota and alter host susceptibility to enteric infections. Here, we demonstrate the effect of L-arabinose on bacterial infection by using a mouse infection model with Salmonella enterica serovar Typhimurium (S. Tm). In the presence of microbiota, L-arabinose induces a dramatic expansion of Enterobacteriaceae, thereby decreasing the microbiota diversity and causing more severe systemic infection. However, L-arabinose supplementation does not alter the disease progression of Salmonella infection in a microbiota-depleted mouse model. More importantly, short-term supplementation of L-arabinose fails to exert anti-diabetic effects in Salmonella-infected hyperglycemia mice and still promotes infection. Overall, our work reveals that a high intake of dietary L-arabinose supports a bloom of Enterobacteriaceae in Salmonella-infected gut, further accelerating the process of systemic infection.IMPORTANCEL-arabinose is a promising natural sweetener and food additive for the regulation of hyperglycemia. Since diabetic subjects are more susceptible to infections, the safety of dietary L-arabinose in diabetic patients experiencing infection remains a concern. Our findings reveal that L-arabinose exacerbates Salmonella infection outcome by inducing gut microbiota dysbiosis in mice. High dietary intake of L-arabinose may be deleterious for diabetic individuals undergoing infection.


Subject(s)
Arabinose , Dysbiosis , Gastrointestinal Microbiome , Salmonella Infections , Salmonella typhimurium , Animals , Gastrointestinal Microbiome/drug effects , Dysbiosis/microbiology , Arabinose/pharmacology , Mice , Salmonella Infections/microbiology , Salmonella typhimurium/drug effects , Mice, Inbred C57BL , Male , Enterobacteriaceae/drug effects
2.
Sci Rep ; 14(1): 11219, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755221

ABSTRACT

Breast cancer patients often have a poor prognosis largely due to lack of effective targeted therapy. It is now well established that monosaccharide enhances growth retardation and chemotherapy sensitivity in tumor cells. We investigated whether D-arabinose has capability to restrict the proliferation of tumor cells and its mechanism. Here, we report that D-arabinose induced cytotoxicity is modulated by autophagy and p38 MAPK signaling pathway in breast cancer cell lines. The proliferation of cells was evaluated by CCK-8 and Colony formation assay. The distribution of cells in cell cycle phases was analyzed by flow cytometry. Cell cycle, autophagy and MAPK signaling related proteins were detected by western blotting. Mouse xenograft model was used to evaluate the efficacy of D-arabinose in vivo. The proliferation of cells was dramatically inhibited by D-arabinose exposure in a dose-dependent manner, which was relevant to cell cycle arrest, as demonstrated by G2/M cell cycle restriction and ectopic expression of cell cycle related proteins. Mechanistically, we further identified that D-arabinose is positively associated with autophagy and the activation of the p38 MAPK signaling in breast cancer. In contrast, 3-Ma or SB203580, the inhibitor of autophagy or p38 MAPK, reversed the efficacy of D-arabinose. Additionally, D-arabinose in vivo treatment could significantly inhibit xenograft growth of breast cancer cells. Our findings were the first to reveal that D-arabinose triggered cell cycle arrest by inducing autophagy through the activation of p38 MAPK signaling pathway in breast cancer cells.


Subject(s)
Arabinose , Autophagy , Breast Neoplasms , Cell Cycle Checkpoints , Cell Proliferation , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases , Autophagy/drug effects , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Animals , Female , p38 Mitogen-Activated Protein Kinases/metabolism , Mice , Arabinose/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , Xenograft Model Antitumor Assays , Mice, Nude , Mice, Inbred BALB C
3.
Food Funct ; 15(9): 5073-5087, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38656276

ABSTRACT

L-Arabinose, lactulose, and Lactobacillus plantarum (L. plantarum) have been reported to have glucolipid-lowering effects. Here, the effects of L-arabinose and lactulose combined with L. plantarum on obesity traits were investigated. According to the experimental results, the combination of L-arabinose, lactulose, and L. plantarum was more effective at reducing body weight, regulating glucolipid metabolism, and improving insulin resistance. Besides, this combination showed immunomodulatory activity by adjusting the T lymphocyte subsets and reduced the immune-related cytokine production. Moreover, it improved the gut barrier, ameliorated the disorder of gut microbiota, and upregulated the levels of SCFAs. More importantly, the AL group, LP group, and ALLP group showed different regulatory effects on the abundance of Bifidobacterium and Lactobacillus due to the presence of lactulose and L. plantarum. These findings elucidate that the combination of L-arabinose, lactulose, and L. plantarum constitutes a new synbiotic combination to control obesity by modulating glucolipid metabolism, immunomodulatory activity, inflammation, gut barrier, gut microbiota and production of SCFAs.


Subject(s)
Arabinose , Diet, High-Fat , Gastrointestinal Microbiome , Lactobacillus plantarum , Lactulose , Mice, Inbred C57BL , Obesity , Animals , Obesity/metabolism , Arabinose/pharmacology , Mice , Lactulose/pharmacology , Male , Gastrointestinal Microbiome/drug effects , Probiotics/pharmacology , Probiotics/administration & dosage , Insulin Resistance
4.
J Glob Antimicrob Resist ; 37: 108-121, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38552872

ABSTRACT

OBJECTIVES: A concern with the ESKAPE pathogen, Enterobacter bugandensis, and other species of the Enterobacter cloacae complex, is the frequent appearance of multidrug resistance against last-resort antibiotics, such as polymyxins. METHODS: Here, we investigated the responses to polymyxin B (PMB) in two PMB-resistant E. bugandensis clinical isolates by global transcriptomics and deletion mutagenesis. RESULTS: In both isolates, the genes of the CrrAB-regulated operon, including crrC and kexD, displayed the highest levels of upregulation in response to PMB. ∆crrC and ∆kexD mutants became highly susceptible to PMB and lost the heteroresistant phenotype. Conversely, heterologous expression of CrrC and KexD proteins increased PMB resistance in a sensitive Enterobacter ludwigii clinical isolate and in the Escherichia coli K12 strain, W3110. The efflux pump, AcrABTolC, and the two component regulators, PhoPQ and CrrAB, also contributed to PMB resistance and heteroresistance. Additionally, the lipid A modification with 4-L-aminoarabinose (L-Ara4N), mediated by the arnBCADTEF operon, was critical to determine PMB resistance. Biochemical experiments, supported by mass spectrometry and structural modelling, indicated that CrrC is an inner membrane protein that interacts with the membrane domain of the KexD pump. Similar interactions were modeled for AcrB and AcrD efflux pumps. CONCLUSION: Our results support a model where drug efflux potentiated by CrrC interaction with membrane domains of major efflux pumps combined with resistance to PMB entry by the L-Ara4N lipid A modification, under the control of PhoPQ and CrrAB, confers the bacterium high-level resistance and heteroresistance to PMB.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Enterobacter , Lipid A , Microbial Sensitivity Tests , Polymyxin B , Polymyxin B/pharmacology , Enterobacter/genetics , Enterobacter/drug effects , Enterobacter/metabolism , Anti-Bacterial Agents/pharmacology , Lipid A/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Arabinose/metabolism , Arabinose/pharmacology , Arabinose/analogs & derivatives , Humans , Gene Expression Regulation, Bacterial , Operon , Drug Resistance, Multiple, Bacterial/genetics , Enterobacteriaceae Infections/microbiology , Drug Resistance, Bacterial , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism
5.
Pharmacol Res ; 202: 107136, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38460778

ABSTRACT

CREB-regulated transcription coactivator 1 (CRTC1), a pivotal synaptonuclear messenger, regulates synaptic plasticity and transmission to prevent depression. Despite exhaustive investigations into CRTC1 mRNA reductions in the depressed mice, the regulatory mechanisms governing its transcription remain elusive. Consequently, exploring rapid but non-toxic CRTC1 inducers at the transcriptional level is important for resisting depression. Here, we demonstrate the potential of D-arabinose, a unique monosaccharide prevalent in edible-medicinal plants, to rapidly enter the brain and induce CRTC1 expression, thereby eliciting rapid-acting and persistent antidepressant responses in chronic restrain stress (CRS)-induced depressed mice. Mechanistically, D-arabinose induces the expressions of peroxisome proliferator-activated receptor gamma (PPARγ) and transcription factor EB (TFEB), thereby activating CRTC1 transcription. Notably, we elucidate the pivotal role of the acetyl-CoA synthetase short-chain family member 2 (ACSS2) as an obligatory mediator for PPARγ and TFEB to potentiate CRTC1 transcription. Furthermore, D-arabinose augments ACSS2-dependent CRTC1 transcription by activating AMPK through lysosomal AXIN-LKB1 pathway. Correspondingly, the hippocampal down-regulations of ACSS2, PPARγ or TFEB alone failed to reverse CRTC1 reductions in CRS-exposure mice, ultimately abolishing the anti-depressant efficacy of D-arabinose. In summary, our study unveils a previously unexplored role of D-arabinose in activating the ACSS2-PPARγ/TFEB-CRTC1 axis, presenting it as a promising avenue for the prevention and treatment of depression.


Subject(s)
Arabinose , PPAR gamma , Mice , Animals , PPAR gamma/genetics , PPAR gamma/metabolism , Arabinose/pharmacology , Arabinose/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain/metabolism
6.
Int Immunopharmacol ; 126: 111188, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37995573

ABSTRACT

There is a growing amount of research that highlights the significant involvement of metabolic imbalance and the inflammatory response in the advancement of colitis. Arabinose is a naturally occurring bioactive monosaccharide that plays a crucial role in the metabolic processes and synthesis of many compounds in living organisms. However, the more detailed molecular mechanism by which the administration of arabinose alleviates the progression of colitis and its associated carcinogenesis is still not fully understood. In the present study, arabinose is recognized as a significant and inherent protector of the intestinal mucosal barrier through its role in preserving the integrity of tight junctions within the intestines. Also, it is important to note that there is a positive correlation between the severity of inflammatory bowel disease (IBD) and colorectal cancer (CRC), as well as chemically-induced colitis in mice, and lower levels of arabinose in the bloodstream. In two mouse models of colitis, caused by dextran sodium sulfate (DSS) or by spontaneous colitis in IL-10-/- mice, damage to the intestinal mucosa was reduced by giving the mice arabinose. When arabinose is administrated to model with colitis, it sets off a chain of events that help keep the lysosomes together and stop cathepsin B from being released. During the progression of intestinal epithelial injury, this process blocks myosin light chain kinase (MLCK) from damaging tight junctions and causing mitochondrial dysfunction. In summary, the results of the study have provided evidence supporting the beneficial effects of arabinose in mitigating the progression of colitis. This is achieved through its ability to avoid dysregulation of the intestinal barrier. Consequently, arabinose may hold promise as a therapeutic supplementation for the management of colitis.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Mice , Animals , Arabinose/therapeutic use , Arabinose/metabolism , Arabinose/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Tight Junctions , Intestinal Mucosa , Dextran Sulfate/pharmacology , Disease Models, Animal , Mice, Inbred C57BL
7.
Cell Chem Biol ; 30(11): 1366-1376.e7, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37536341

ABSTRACT

Stimulator of interferon genes (STING) agonists are promising candidates for vaccine adjuvants and antitumor immune stimulants. The most potent natural agonist of STING, 2',3'-cyclic GMP-AMP (2',3'-cGAMP), is subject to nuclease-mediated inherent metabolic instability, thereby placing limits on its clinical efficacy. Here, we report on a new class of chemically synthesized sugar-modified analogs of 2',3'-cGAMP containing arabinose and xylose sugar derivatives that bind mouse and human STING alleles with high affinity. The co-crystal structures demonstrate that such analogs act as 2',3'-cGAMP mimetics that induce the "closed" conformation of human STING. These analogs show significant resistance to hydrolysis mediated by ENPP1 and increased stability in human serum, while retaining similar potency as 2',3'-cGAMP at inducing IFN-ß secretion from human THP1 cells. The arabinose- and xylose-modified 2',3'-cGAMP analogs open a new strategy for overcoming the inherent nuclease-mediated vulnerability of natural ribose cyclic nucleotides, with the additional benefit of high translational potential as cancer therapeutics and vaccine adjuvants.


Subject(s)
Arabinose , Xylose , Humans , Animals , Mice , Arabinose/pharmacology , Adjuvants, Vaccine , Nucleotides, Cyclic/metabolism
8.
Nutrition ; 111: 112041, 2023 07.
Article in English | MEDLINE | ID: mdl-37207566

ABSTRACT

OBJECTIVES: The global prevalence of obesity, a chronically trophic metabolic disease, has garnered significant attention. The aim of this study was to investigate L-arabinose, a unique functional sugar that improves insulin resistance and intestinal environment while promoting probiotic proliferation, for its potential in preventing obesity induced by a high-fat and high-sugar (HFHS) diet in mice. METHODS: The L-arabinose group was intragastrically administered with 0.4 mL 60 mg/(kg body weight) L-arabinose for 8 wk. The metformin group was intragastrically administered at 0.4 mL 300 mg/(kg body weight), as a positive control group. RESULTS: Treatment with L-arabinose resulted in a reduction of various obesity symptoms, such as prevented weight gain, increased liver-to-body ratio, decreased insulin, homeostasis model assessment for insulin resistance (HOMA-IR) index, and lipopolysaccharide (LPS) levels, as well as improved insulin resistance, reduced fat volume, inhibited hepatic steatosis, and repaired the pancreas. The L-arabinose treatment also improved lipid metabolism and inflammatory response, decreased the Firmicutes-to-Bacteroidetes ratio at the phylum level, and increased the relative abundance of Parabacteroides gordonii and Akkermansia muciniphila at the species level. CONCLUSION: Based on these results, L-arabinose could be a promising candidate for combating obesity and obesity-related diseases by regulating insulin resistance and gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Insulin Resistance , Mice , Animals , Arabinose/pharmacology , Mice, Obese , Diet, High-Fat/adverse effects , Obesity/metabolism , Mice, Inbred C57BL
9.
Nutrients ; 15(7)2023 Mar 25.
Article in English | MEDLINE | ID: mdl-37049441

ABSTRACT

Rare sugars have recently attracted attention as potential sugar replacers. Understanding the biochemical and biological behavior of these sugars is of importance in (novel) food formulations and prevention of type 2 diabetes. In this study, we investigated whether rare sugars may positively affect intestinal and liver metabolism, as well as muscle insulin sensitivity, compared to conventional sugars. Rare disaccharide digestibility, hepatic metabolism of monosaccharides (respirometry) and the effects of sugars on skeletal muscle insulin sensitivity (impaired glucose uptake) were investigated in, respectively, Caco-2, HepG2 and L6 cells or a triple coculture model with these cells. Glucose and fructose, but not l-arabinose, acutely increased extracellular acidification rate (ECAR) responses in HepG2 cells and impaired glucose uptake in L6 cells following a 24 h exposure at 28 mM. Cellular bioenergetics and digestion experiments with Caco-2 cells indicate that especially trehalose (α1-1α), D-Glc-α1,2-D-Gal, D-Glc-α1,2-D-Rib and D-Glc-α1,3-L-Ara experience delayed digestion and reduced cellular impact compared to maltose (α1-4), without differences on insulin-stimulated glucose uptake in a short-term setup with a Caco-2/HepG2/L6 triple coculture. These results suggest a potential for l-arabinose and specific rare disaccharides to improve metabolic health; however, additional in vivo research with longer sugar exposures should confirm their beneficial impact on insulin sensitivity in humans.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Diabetes Mellitus, Type 2/metabolism , Caco-2 Cells , Arabinose/pharmacology , Arabinose/metabolism , Glucose/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , Liver/metabolism , Disaccharides/pharmacology
10.
J Mol Recognit ; 36(1): e2993, 2023 01.
Article in English | MEDLINE | ID: mdl-36112092

ABSTRACT

Atomic force microscopy (AFM) was used to conduct single-molecule imaging of protein/DNA complexes involved in the regulation of the arabinose operon of Escherichia coli. In the presence of arabinose, the transcription regulatory protein AraC binds to a 38 bp region consisting of the araI1 and araI2 half-sites. The domain positioning of full-length AraC, when bound to DNA, was not previously known. In this study, AraC was combined with 302 and 560 bp DNA and arabinose, deposited on a mica substrate, and imaged with AFM in air. High resolution images of 560 bp DNA, where bound protein was visible, showed that AraC induces a bend in the DNA with an angle 60° ± 12° with a median of 55°. These results are consistent with earlier gel electrophoresis measurements that measured the DNA bend angle based on migration rates. By using known domain structures of AraC, geometric constraints, and contacts determined from biochemical experiments, we developed a model of the tertiary and quaternary structure of DNA-bound AraC in the presence of arabinose. The DNA bend angle predicted by the model is in agreement with the measurement values. We discuss the results in view of other regulatory proteins that cause DNA bending and formation of the open complex to initiate transcription.


Subject(s)
AraC Transcription Factor , Escherichia coli Proteins , AraC Transcription Factor/genetics , AraC Transcription Factor/chemistry , AraC Transcription Factor/metabolism , Escherichia coli Proteins/metabolism , Microscopy, Atomic Force , Cytarabine/metabolism , Repressor Proteins/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Bacterial Proteins/metabolism , Arabinose/chemistry , Arabinose/metabolism , Arabinose/pharmacology , Transcription Factors/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , DNA/metabolism , Protein Binding
11.
Food Funct ; 13(19): 10158-10170, 2022 Oct 03.
Article in English | MEDLINE | ID: mdl-36106930

ABSTRACT

Oat ß-glucan (OBG) and L-arabinose (LA) have exhibited positive effects on diabetes and its complications. However, it is unclear whether OBG and LA have a synergistic effect. We investigated the effect of variable compositions (OBG : LA = 1 : 1, 1 : 2, 1 : 4,1 : 6, 1 : 8, 1 : 10, 2 : 1, 4 : 1, 6 : 1, 8 : 1, 10 : 1) on glucose uptake in IR-HepG2 cells induced by dexamethasone (DEX) to find out the optimal composition showing synergistic effects. Furthermore, this study evaluated the anti-diabetic activity of the optimal composition in db/db mice. In vitro, the OBG : LA = 1 : 1 group showed the strongest synergistic effects among the varied compositions, outperforming OBG and LA alone. In vivo, there were more beneficial effects in the OBG : LA = 1 : 1 group compared with the OBG and LA single-dosing groups. OBG : LA = 1 : 1 supplementation markedly decreased the levels of fasting blood glucose (FBG) and insulin (INS) in serum, improved glucose tolerance and insulin sensitivity, lowered blood lipid levels, and reduced liver lipid accumulation. Moreover, the western blot results indicated that the OBG : LA = 1 : 1 group up-regulated the protein expression of glucose transporter-4 (GLUT4), phosphatidylinositol 3-kinase (PI3K), and phospho-protein kinase B (p-AKT), while down-regulating the protein expression of phospho-phosphorylated insulin receptor substrate-1 (p-IRS1) to enhance insulin transduction in liver tissues. These findings suggest that OBG : LA = 1 : 1 synergistically ameliorated glucose metabolism disorders and alleviated insulin resistance by promoting the PI3K/AKT pathway in the liver.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Arabinose/pharmacology , Blood Glucose/metabolism , Dexamethasone/pharmacology , Diabetes Mellitus, Type 2/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Hep G2 Cells , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mice , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , beta-Glucans
12.
Food Funct ; 13(18): 9268-9284, 2022 Sep 22.
Article in English | MEDLINE | ID: mdl-35993148

ABSTRACT

The effect of different extraction processes on the physicochemical characterization, digestibility, antioxidant activity and prebiotic activity of Isaria cicadae Miquel (ICM) fruiting body polysaccharides was studied. Furthermore, the effect of ultrasound-assisted extraction of ICM (U-ICM) on gut microbiota, the intestinal barrier and immune response was deeply explored. This study found that ICMs showed high indigestibility in both α-amylase and artificial gastric juice, indicating that ICMs have the potential as dietary fiber. In contrast, U-ICM had the best antioxidant activity and prebiotic potential. Meanwhile, there was a structure-activity relationship between the antioxidant activity of ICMs and the content of uronic acid, arabinose and galactose. When healthy mice were fed U-ICM for 42 days, the relative abundances of Lactobacillus, Akkermansia, and Bacteroides were found to increase significantly, while that of Clostridium decreased significantly. Meanwhile, U-ICM significantly promotes the expression of tight junction protein and the production of cytokines, indicating that U-ICM had the function of enhancing the intestinal barrier and regulating the host immune response. In conclusion, U-ICM as dietary fiber has the potential to be developed as a gut health-promoting prebiotic component or functional food. This research provided a valuable resource for further exploring the structure-activity relationship and prebiotic activity of ICMs.


Subject(s)
Gastrointestinal Microbiome , Animals , Antioxidants/pharmacology , Arabinose/pharmacology , Cordyceps , Cytokines/pharmacology , Dietary Fiber/pharmacology , Galactose/pharmacology , Immunity , Mice , Polysaccharides/chemistry , Polysaccharides/pharmacology , Tight Junction Proteins , Uronic Acids/pharmacology , alpha-Amylases/pharmacology
13.
Int J Biol Macromol ; 220: 638-658, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35973483

ABSTRACT

The study aims to investigate the constituents, adjuvant effects, and underlying mechanisms of purified polysaccharides from cultivated Cistanche deserticola (C. deserticola). Two macromolecules designated as CCDP-1 (26.5 kDa) and CCDP-2 (32.3 kDa) from C. deserticola were respectively identified as carbohydrate-lignin complexes with 44.1 % and 43.8 % lignin. CCDP-1 and CCDP-2 were composed of glucose, rhamnose, galactose, arabinose, and mannose respectively in the molar ratios of 7.22: 5.98:2.51:1.81:1.00 and 6.57:8.48:4.20:2.72:1.00. An in vitro experiment revealed that endotoxin-free CCDP-1 and CCDP-2 promoted splenocyte proliferation without cytotoxicity, but CCDP-2 induced dendritic cell (DC) maturation more efficiently than CCDP-1. An in vivo experiment suggested that CCDP-2 enhanced OVA-specific antibody production, antigen-specific T-cell activation, IFN-γ production, IL-4 production, and DC activation. Notably, CCDP-2 elicited a Th1-biased response. Mechanically, CCDP-2 upregulated CD40, CD80, CD86, and MHC II, facilitated allogeneic T-cell proliferation and Th1/Th2 cytokines, improved IFN-γ, IL-12, IL-6, and TNF-α production, and decreased endocytosis from DCs in vitro. Blocking assays indicated that TLR2 and TLR4 were the membrane receptor candidates of DCs. Western blot implied that CCDP-2 with the immune-enhancing activities were involved in the activation of MAPKs and NF-κB pathways in a dose-/time-related manner and could be employed as a more balanced Th1/Th2 adjuvant for vaccine exploitation.


Subject(s)
Cistanche , Vaccines , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Arabinose/pharmacology , Cistanche/chemistry , Cytokines/metabolism , Dendritic Cells , Galactose/metabolism , Glucose/metabolism , Interleukin-12/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Lignin/metabolism , Mannose/metabolism , NF-kappa B/metabolism , Polysaccharides/chemistry , Rhamnose/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vaccines/pharmacology
14.
Cell Rep ; 40(3): 111087, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858544

ABSTRACT

Microbiota-accessible carbohydrates (MACs) exert health-promoting effects, but how each MAC impacts gut microbiota and regulates host physiology remains unclear. Here, we show that l-arabinose and sucrose cooperatively act on gut microbiota and exert anti-obesogenic effects. Specifically, l-arabinose, a monosaccharide that is poorly absorbed in the gut and inhibits intestinal sucrase, suppresses diet-induced obesity in mice in the presence of sucrose. Additionally, the suppressive effect of l-arabinose on adiposity is abrogated in mice lacking the short-chain fatty acid (SCFA) receptors GPR43 and GPR41. Mechanistically, l-arabinose increases the relative abundance of acetate and propionate producers (e.g., Bacteroides), while sucrose enhances SCFA production. Furthermore, l-arabinose and sucrose activate the glycolytic and pentose phosphate pathways of Bacteroides, respectively, indicating that they synergistically promote acetate production through distinct pathways. These findings suggest that each MAC has a unique property and thus may serve as a precision gut-microbiota modulator to promote host homeostasis.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Arabinose/pharmacology , Bacteroides/metabolism , Carbohydrates , Fatty Acids, Volatile/metabolism , Mice , Obesity/metabolism , Sucrose
15.
Anaerobe ; 75: 102533, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35143955

ABSTRACT

OBJECTIVES: Biofilm formation on dental implant surfaces can cause peri-implant mucositis and peri-implantitis. Lectins are involved in interactions between bacteria or between bacteria and their hosts. Disrupting these interactions via specific sugars can result in reduced adhesion and biofilm formation. The purpose of this study was to identify sugars that function as antiadhesion or antibiofilm agents on titanium discs. METHODS: Of the sugars tested, the sugars that did not affect the planktonic growth of Streptococcus oralis, Fusobacterium nucleatum, and Porphyromonas gingivalis were selected. The selected sugars were assessed for their ability to inhibit biofilm formation of bacteria in single and consortium species by crystal violet staining, confocal laser scanning microscopy after live/dead staining, and scanning electron microscopy. The sugars were evaluated for their ability to inhibit activity of the quorum sensing molecule autoinducer 2 (AI-2) by bioluminescence assay. RESULTS: Biofilm formation of single bacteria or consortia of S. oralis, F. nucleatum, and P. gingivalis on titanium discs was significantly inhibited in the presence of d-arabinose. Pretreating titanium discs with d-arabinose for 3 min inhibited biofilm formation at a level comparable to that observed when d-arabinose was present over the entire period, suggesting that d-arabinose had initial anti-adhesive activity. In addition, d-arabinose inhibited the activity of AI-2. CONCLUSIONS: d-Arabinose may be a good candidate for application as an antibiofilm agent and AI-2 inhibitor.


Subject(s)
Peri-Implantitis , Titanium , Arabinose/pharmacology , Biofilms , Fusobacterium nucleatum , Humans , Porphyromonas gingivalis , Titanium/pharmacology
16.
Br J Nutr ; 128(6): 1072-1081, 2022 09 28.
Article in English | MEDLINE | ID: mdl-34657640

ABSTRACT

Dietary interventions to delay carbohydrate digestion or absorption can effectively prevent hyperglycaemia in the early postprandial phase. L-arabinose can specifically inhibit sucrase. It remains to be assessed whether co-ingestion of L-arabinose with sucrose delays sucrose digestion, attenuates subsequent glucose absorption and impacts hepatic glucose output. In this double-blind, randomised crossover study, we assessed blood glucose kinetics following ingestion of a 200-ml drink containing 50 g of sucrose with 7·5 g of L-arabinose (L-ARA) or without L-arabinose (CONT) in twelve young, healthy participants (24 ± 1 years; BMI: 22·2 ± 0·5 kg/m2). Plasma glucose kinetics were determined by a dual stable isotope methodology involving ingestion of (U-13C6)-glucose-enriched sucrose, and continuous intravenous infusion of (6,6-2H2)-glucose. Peak glucose concentrations reached 8·18 ± 0·29 mmol/l for CONT 30 min after ingestion. In contrast, the postprandial rise in plasma glucose was attenuated for L-ARA, because peak glucose concentrations reached 6·62 ± 0·18 mmol/l only 60 min after ingestion. The rate of exogenous glucose appearance for L-ARA was 67 and 57 % lower compared with CONT at t = 15 min and 30 min, respectively, whereas it was 214 % higher at t = 150 min, indicating a more stable absorption of exogenous glucose for L-ARA compared with CONT. Total glucose disappearance during the first hour was lower for L-ARA compared with CONT (11 ± 1 v. 17 ± 1 g, P < 0·0001). Endogenous glucose production was not differentially affected at any time point (P = 0·27). Co-ingestion of L-arabinose with sucrose delays sucrose digestion, resulting in a slower absorption of sucrose-derived glucose without causing adverse effects in young, healthy adults.


Subject(s)
Blood Glucose , Glucose , Male , Adult , Humans , Female , Arabinose/pharmacology , Cross-Over Studies , Sucrose , Insulin , Eating , Postprandial Period
17.
Neurosci Lett ; 764: 136205, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34478818

ABSTRACT

Lactate transport is an important means of communication between astrocytes and neurons and is implicated in a variety of neurobiological processes. However, the connection between astrocyte-neuron lactate transport and nociceptive modulation has not been well established. Here, we found that Complete Freund's adjuvant (CFA)-induced inflammation pain leads to a significant increase in extracellular lactate levels in the anterior cingulate cortex (ACC). Inhibition of glycogenolysis and lactate release in the ACC disrupted the persistent, but not acute, inflammation pain induced by CFA, and this effect was reversed by exogenous L-lactate administration. Knocking down the expression of lactate transporters (MCT1, MCT4, or MCT2) also disrupted the long lasting inflammation pain induced by CFA. Moreover, glycogenolysis in the ACC is critical for the induction of molecular changes related to neuronal plasticity, including the induction of phospho- (p-) ERK, p-CREB, and Fos. Taken together, our findings indicate that astrocyte-neuron lactate transport in the ACC is critical for the occurrence of persistent inflammation pain, suggesting a novel mechanism underlying chronic pain.


Subject(s)
Arabinose/pharmacology , Cell Communication/immunology , Chronic Pain/immunology , Gyrus Cinguli/pathology , Imino Furanoses/pharmacology , Lactic Acid/metabolism , Sugar Alcohols/pharmacology , Animals , Arabinose/therapeutic use , Astrocytes/metabolism , Cell Communication/drug effects , Chronic Pain/drug therapy , Chronic Pain/pathology , Disease Models, Animal , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Glycogenolysis/drug effects , Glycogenolysis/immunology , Gyrus Cinguli/cytology , Gyrus Cinguli/drug effects , Gyrus Cinguli/immunology , Humans , Imino Furanoses/therapeutic use , Male , Mice , Neuronal Plasticity/drug effects , Neuronal Plasticity/immunology , Neurons/metabolism , Sugar Alcohols/therapeutic use
18.
Int J Biol Macromol ; 183: 2074-2087, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34097961

ABSTRACT

Lycium barbarum polysaccharides (LBPs) are known for their beneficial effects on diabetes, NAFLD and related chronic metabolic diseases induced by high-fat diet (HFD). However, the relevant researches are mainly about the whole crude polysaccharides, the specific active ingredient of LBPs and its bioactivity have been rarely explored. Herein, a homogeneous polysaccharide (LBP-W) was isolated and purified from crude LBPs. Structure characterizations indicated that LBP-W contained a main chain consisting of a repeated unit of →6)-ß-Galp(1 â†’ residues with branches composed of α-Araf, ß-Galp and α-Rhap residues at position C-3. The objective of this study was to evaluate the anti-obesogenic effect of LBP-W and figure out the underlying mechanisms. In vivo efficacy trial illustrated that LBP-W supplements can alleviate HFD-induced mice obesity significantly. Gut microbiota analysis showed that LBP-W not only improved community diversity of intestinal flora, but also regulated their specific genera. Moreover, LBP-W can increase the content of short-chain fatty acids (SCFAs), a metabolite of the intestinal flora. In summary, all these results demonstrated that the homogeneous polysaccharide purified from L. barbarum could be used as a prebiotic agent to improve obesity by modulating the composition of intestinal flora and the metabolism of SCFAs.


Subject(s)
Anti-Obesity Agents/pharmacology , Bacteria/drug effects , Drugs, Chinese Herbal/pharmacology , Energy Metabolism/drug effects , Gastrointestinal Microbiome/drug effects , Obesity/drug therapy , Prebiotics , Animals , Anti-Obesity Agents/chemistry , Arabinose/chemistry , Arabinose/pharmacology , Bacteria/growth & development , Bacteria/metabolism , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Dysbiosis , Fatty Acids/blood , Galactose/chemistry , Galactose/pharmacology , Male , Mice, Inbred C57BL , Molecular Structure , Obesity/blood , Obesity/microbiology , Rhamnose/chemistry , Rhamnose/pharmacology , Structure-Activity Relationship
19.
Carbohydr Res ; 503: 108311, 2021 May.
Article in English | MEDLINE | ID: mdl-33866267

ABSTRACT

A series of oleanolic acid derivatives bearing acetyl-substituted l-arabinose moiety has been synthesized and screened in vitro for cytotoxicity against ten cancer cell lines and four normal cell lines. The antiproliferative evaluation indicated that synthetic derivatives showed excellent selectivity, as they were toxic against only A431 cell line. Among them, the compound 6 possesses the best inhibitory activity. A series of pharmacology experiments showed that compound 6 significantly induced A431 cells apoptosis and cell cycle arrest, which could serve as a promising lead candidate for further study.


Subject(s)
Antineoplastic Agents/pharmacology , Arabinose/pharmacology , Oleanolic Acid/pharmacology , Saponins/pharmacology , Acetylation , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Arabinose/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Conformation , Oleanolic Acid/chemical synthesis , Oleanolic Acid/chemistry , Saponins/chemical synthesis , Saponins/chemistry
20.
Biochem Biophys Res Commun ; 556: 163-170, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33845307

ABSTRACT

Although efficient methods of gene silencing have been established in eukaryotes, many different techniques are still used in bacteria due to the lack of a standardized tool. Here, we developed a convenient and efficient method to downregulate the expression of a specific gene using ∼140 nucleotide RNA with a 24-nucleotide antisense region from an arabinose-inducible expression plasmid by taking Escherichia coli lacZ and phoA genes encoding ß-galactosidase and alkaline phosphatase, respectively, as target genes to evaluate the model. We examined the antisense RNA (asRNA) design, including targeting position, uORF stability elements at the 5'-end, and Hfq-binding module at the 3'-end, and inducer amount required to obtain effective experimental conditions for gene silencing. Furthermore, we constructed multiplexed dual-acting asRNA genes in the plasmid, which were transcribed as polycistronic RNA and were able to knockdown multiple target genes simultaneously. We observed the highest inhibition level of 98.6% when lacZ was targeted using the pMKN104 asRNA expression plasmid, containing a five times stronger PBAD -10 promoter sequence with no requirement of the Hfq protein for repression. These features allow the system to be utilized as an asRNA expression platform in many bacteria, besides E. coli, for gene regulation.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Gene Knockdown Techniques/methods , Gene Silencing , Genes/genetics , RNA, Antisense/genetics , Arabinose/metabolism , Arabinose/pharmacology , Base Sequence , Codon, Initiator/genetics , Escherichia coli/drug effects , Gene Expression Regulation, Bacterial/drug effects , Gene Silencing/drug effects , Genes/drug effects , Genes, Reporter , Plasmids/drug effects , Plasmids/genetics , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA, Antisense/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL