Home>>Signaling Pathways>> Proteases>> Endogenous Metabolite>>D-Arabinose

D-Arabinose Sale

(Synonyms: D-阿拉伯糖) 目录号 : GC38700

A monosaccharide

D-Arabinose Chemical Structure

Cas No.:28697-53-2

规格 价格 库存 购买数量
100mg
¥450.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

产品文档

Quality Control & SDS

View current batch:

产品描述

D-Arabinose is a monosaccharide. It has been found in the O-specific polysaccharide of LPS in P. maltophilia.1 D-Arabinose (50 and 100 mM) inhibits S. oralis, F. nucleatum, and P. gingivalis biofilm formation as well as the activity of autoinducer 2, a quorum-sensing molecule involved in the pathogenesis of periodontitis, in V. harveyi in a reporter assay.2 It reduces the growth of C. elegans (IC50 = 7.5 mM), an effect that can be reversed by D-ribose or D-fructose, but not D-glucose.3

1.Wilkinson, S.G., Galbraith, L., and Anderton, W.J.Lipopolysaccharides from Pseudomonas maltophilia: Composition of the lipopolysaccharide and structure of the side-chain polysaccharide from strain N.C.I.B. 9204Carbohydr. Res.112(2)241-252(1983) 2.An, S.-J., Namkung, J.-U., Ha, K.-W., et al.Inhibitory effect of ?-arabinose on oral bacteria biofilm formation on titanium discsAnaerobe102533(2022) 3.Sakoguchi, H., Yoshihara, A., Shintani, T., et al.Growth inhibitory effect of ?-arabinose against the nematode Caenorhabditis elegans: Discovery of a novel bioactive monosaccharideBioorg. Med. Chem. Lett.26(3)726-729(2106)

Chemical Properties

Cas No. 28697-53-2 SDF
别名 D-阿拉伯糖
Canonical SMILES OC1[C@H]([C@@H]([C@@H](CO1)O)O)O
分子式 C5H10O5 分子量 150.13
溶解度 DMSO : 30mg/mL 储存条件 Store at RT
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 6.6609 mL 33.3045 mL 66.6089 mL
5 mM 1.3322 mL 6.6609 mL 13.3218 mL
10 mM 0.6661 mL 3.3304 mL 6.6609 mL
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

A review on selective l-fucose/D-Arabinose isomerases for biocatalytic production of l-fuculose/d-ribulose

Int J Biol Macromol 2021 Jan 31;168:558-571.PMID:33296692DOI:10.1016/j.ijbiomac.2020.12.021.

L-Fuculose and D-ribulose are kinds of rare sugars used in food, agriculture, and medicine industries. These are pentoses and categorized into the two main groups, aldo pentoses and ketopentoses. There are 8 aldo- and 4 ketopentoses and only fewer are natural, while others are rare sugars found in a very small amount in nature. These sugars have great commercial applications, especially in many kinds of drugs in the medicine industry. The synthesis of these sugars is very expensive, difficult by chemical methods due to its absence in nature, and could not meet industry demands. The pentose izumoring strategy offers a complete enzymatic tactic to link all kinds of pentoses using different enzymes. The enzymatic production of L-fuculose and D-ribulose through L-fucose isomerase (L-FI) and D-Arabinose isomerase (D-AI) is the inexpensive and uncomplicated method up till now. Both enzymes have similar kinds of isomerizing mechanisms and each enzyme can catalyze both L-fucose and D-Arabinose. In this review article, the enzymatic process of biochemically characterized L-FI & D-AI, their application to produce L-fuculose and D-ribulose and its uses in food, agriculture, and medicine industries are reviewed.

A new synthesis of d-lyxose from D-Arabinose

Carbohydr Res 2023 Apr;526:108782.PMID:37001234DOI:10.1016/j.carres.2023.108782.

A new synthesis of rare d-lyxose from easily available D-Arabinose is disclosed. The route includes 7 steps with a total 40% yield. Inversion of configuration at C3 promoted by DAST reagent is utilized on trans-2,3-di-hydroxy pentofuranose to provide cis-2,3-di-hydroxy pentofuranose, which is hardly synthesized using normal method.

Biosynthesis of D-Arabinose in mycobacteria - a novel bacterial pathway with implications for antimycobacterial therapy

FEBS J 2008 Jun;275(11):2691-711.PMID:18422659DOI:10.1111/j.1742-4658.2008.06395.x.

Decaprenyl-phospho-arabinose (beta-D-arabinofuranosyl-1-O-monophosphodecaprenol), the only known donor of D-Arabinose in bacteria, and its precursor, decaprenyl-phospho-ribose (beta-D-ribofuranosyl-1-O-monophosphodecaprenol), were first described in 1992. En route to D-arabinofuranose, the decaprenyl-phospho-ribose 2'-epimerase converts decaprenyl-phospho-ribose to decaprenyl-phospho-arabinose, which is a substrate for arabinosyltransferases in the synthesis of the cell-wall arabinogalactan and lipoarabinomannan polysaccharides of mycobacteria. The first step of the proposed decaprenyl-phospho-arabinose biosynthesis pathway in Mycobacterium tuberculosis and related actinobacteria is the formation of D-ribose 5-phosphate from sedoheptulose 7-phosphate, catalysed by the Rv1449 transketolase, and/or the isomerization of d-ribulose 5-phosphate, catalysed by the Rv2465 d-ribose 5-phosphate isomerase. d-Ribose 5-phosphate is a substrate for the Rv1017 phosphoribosyl pyrophosphate synthetase which forms 5-phosphoribosyl 1-pyrophosphate (PRPP). The activated 5-phosphoribofuranosyl residue of PRPP is transferred by the Rv3806 5-phosphoribosyltransferase to decaprenyl phosphate, thus forming 5'-phosphoribosyl-monophospho-decaprenol. The dephosphorylation of 5'-phosphoribosyl-monophospho-decaprenol to decaprenyl-phospho-ribose by the putative Rv3807 phospholipid phosphatase is the committed step of the pathway. A subsequent 2'-epimerization of decaprenyl-phospho-ribose by the heteromeric Rv3790/Rv3791 2'-epimerase leads to the formation of the decaprenyl-phospho-arabinose precursor for the synthesis of the cell-wall arabinans in Actinomycetales. The mycobacterial 2'-epimerase Rv3790 subunit is similar to the fungal D-arabinono-1,4-lactone oxidase, the last enzyme in the biosynthesis of D-erythroascorbic acid, thus pointing to an evolutionary link between the D-arabinofuranose- and L-ascorbic acid-related pathways. Decaprenyl-phospho-arabinose has been a lead compound for the chemical synthesis of substrates for mycobacterial arabinosyltransferases and of new inhibitors and potential antituberculosis drugs. The peculiar (omega,mono-E,octa-Z) configuration of decaprenol has yielded insights into lipid biosynthesis, and has led to the identification of the novel Z-polyprenyl diphosphate synthases of mycobacteria. Mass spectrometric methods were developed for the analysis of anomeric linkages and of dolichol phosphate-related lipids. In the field of immunology, the renaissance in mycobacterial polyisoprenoid research has led to the identification of mimetic mannosyl-beta-1-phosphomycoketides of pathogenic mycobacteria as potent lipid antigens presented by CD1c proteins to human T cells.

The Biosynthesis of D-1,2,4-Butanetriol From D-Arabinose With an Engineered Escherichia coli

Front Bioeng Biotechnol 2022 Mar 24;10:844517.PMID:35402410DOI:10.3389/fbioe.2022.844517.

D-1,2,4-Butanetriol (BT) has attracted much attention for its various applications in energetic materials and the pharmaceutical industry. Here, a synthetic pathway for the biosynthesis of BT from D-Arabinose was constructed and optimized in Escherichia coli. First, E. coli Trans1-T1 was selected for the synthesis of BT. Considering the different performance of the enzymes from different organisms when expressed in E. coli, the synthetic pathway was optimized. After screening two D-Arabinose dehydrogenases (ARAs), two d-arabinonate dehydratases (ADs), four 2-keto acid decarboxylases (ADXs), and three aldehyde reductases (ALRs), ADG from Burkholderia sp., AraD from Sulfolobus solfataricus, KivD from Lactococcus lactis IFPL730, and AdhP from E. coli were selected for the bio-production of BT. After 48 h of catalysis, 0.88 g/L BT was produced by the recombinant strain BT5. Once the enzymes were selected for the pathway, metabolic engineering strategy was conducted for further improvement. The final strain BT5ΔyiaEΔycdWΔyagE produced 1.13 g/L BT after catalyzing for 48 h. Finally, the fermentation conditions and characteristics of BT5ΔyiaEΔycdWΔyagE were also evaluated, and then 2.24 g/L BT was obtained after 48 h of catalysis under the optimized conditions. Our work was the first report on the biosynthesis of BT from D-Arabinose which provided a potential for the large-scale production of d-glucose-based BT.

Inhibitory effect of D-Arabinose on oral bacteria biofilm formation on titanium discs

Anaerobe 2022 Jun;75:102533.PMID:35143955DOI:10.1016/j.anaerobe.2022.102533.

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.