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

D-Allose Sale

(Synonyms: D-阿洛糖) 目录号 : GC62921

D-Allose 是一种内源性代谢产物。

D-Allose Chemical Structure

Cas No.:2595-97-3

规格 价格 库存 购买数量
100 mg
¥540.00
现货
250 mg
¥1,080.00
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-Allose is an endogenous metabolite.

Chemical Properties

Cas No. 2595-97-3 SDF
别名 D-阿洛糖
分子式 C6H12O6 分子量 180.16
溶解度 储存条件 Store at 2-8°C,protect from light
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 5.5506 mL 27.7531 mL 55.5062 mL
5 mM 1.1101 mL 5.5506 mL 11.1012 mL
10 mM 0.5551 mL 2.7753 mL 5.5506 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

D-Allose: molecular pathways and therapeutic capacity in cancer

Curr Mol Pharmacol 2022 Dec 27.PMID:36578261DOI:10.2174/1874467216666221227105011.

Background: Despite the implementation of various cancer therapies, adequate therapeutic efficacy has not been achieved. A growing number of studies have been dedicated to the discovery of new molecules to combat refractory cancer cells efficiently. Recently, the use of a rare type of sugar, D-Allose, has attracted the attention of research communities. In combination with the first-line treatment of cancers, including different types of radiotherapies and chemotherapies, D-Allose has been detected with favorable complementary effects. Understanding the mechanism of therapeutic target molecules will enable us to develop new strategies for cancer patients that do not currently respond to the present therapies. Objective: We aimed to provide a review of the effects of D-Allose in cancer treatment, its mechanisms of action, and gaps in this field that require more investigations. Discussion: With rare exceptions, in many cancer types, including head and neck, lung, liver, bladder, blood, and breast, D-Allose consistently has exhibited anticancer activity in vitro and/or in vivo. Most of the D-Allose functions are mediated through thioredoxin-interacting protein molecules. D-Allose exerts its effects via reactive oxygen species regulation, cell cycle arrest, metabolic reprogramming, autophagy, apoptosis induction, and sensitizing tumors to radiotherapy and chemotherapy. Conclusion: D-Allose has shown great promise for combating tumor cells with no side effects, especially in combination with first-line drugs; however, its potential for cancer therapy has not been comprehensively investigated in vitro or in vivo.

Immunomodulatory effects of D-Allose on cytokine production by plasmacytoid dendritic cells

Biochem Biophys Res Commun 2022 Oct 30;627:130-136.PMID:36030654DOI:10.1016/j.bbrc.2022.08.037.

D-Allose is classified as a 'rare sugar,' i.e., part of the group of monosaccharides that are present in low quantities in the natural world. D-Allose has been demonstrated to exert many physiological functions. The effects of the rare sugars on immune responses are largely unexplored. Here, we investigated the physiological effects of D-Allose on murine dendritic cells' cytokine production. When plasmacytoid dendritic cells (pDCs) were stimulated with a Toll-like receptor 7 (TLR7) ligand, a single-stranded RNA (ssRNA), or a TLR9 ligand, CpG DNA, in the medium containing D-Allose, the productions of both interferon-alpha (IFN-α) and interleukin (IL)-12p40 were severely decreased. In contrast, a normal production of these cytokines was observed when pDCs were stimulated with other TLR7 ligands, an imidazoquinoline, or a guanosine analog. In contrast to the pDCs, conventional dendritic cells (cDCs) produced IL-12p40 and tumor necrosis factor-alpha (TNF-α) in response to an imidazoquinoline or CpG DNA even though D-Allose was present in the medium. D-Allose did not induce pDC death, and not inhibit the endocytic uptake of fluorophore-labeled CpG DNA into pDCs. These results suggested that D-Allose exerts its inhibitory effects after CpG DNA is internalized. We analyzed the TLR7/9 signal-induced activation of downstream signaling molecules in pDCs and observed that when pDCs were stimulated with a ssRNA or CpG DNA, the phosphorylation status of the MAPK family, which includes Erk1/2, JNK/SAPK, and p38 MAPK, was attenuated in the presence of D-Allose compared to D-glucose controls. The stimulation of pDCs with an imidazoquinoline induced a strong phosphorylation of these MAPK family members even in the presence of D-Allose. These findings reveal that D-Allose can inhibit the cytokine production by pDCs stimulated with ssRNA or CpG DNA via an attenuation of the phosphorylation of MAPK family members.

D-Allose alleviates ischemia/reperfusion (I/R) injury in skin flap via MKP-1

Mol Med 2020 Feb 11;26(1):21.PMID:32046628DOI:10.1186/s10020-020-0138-6.

Background: D-Allose was promising in the protection of ischemia/reperfusion (I/R) injury. We intended to investigate the function of D-Allose in skin flap of rat followed by the injury of I/R and whether ERK signal pathway was involved in. Methods: The back flap of Wistar rats was picked up with a vascular bundle of the lateral chest wall. I/R model was made by the venous clamp for 6 h. Rats received D-Allose and PD-98059, the inhibitor of ERK1/2, 30 min before modeling. Morphology of tissue was observed by HE staining. Nitric oxide (NO), myeloperoxidase (MPO), malondialdehyde (MDA) and superoxide dismutase (SOD) levels in skin flap were determined by ELISA kits. mRNA and protein levels were determined by qPCR and Western blot respectively. Results: D-Allose alleviated the condition of pathological changes and raised the survival rate of skin flap injured by I/R. Moreover, D-Allose suppressed NO, MPO and MDA while elevated SOD levels during I/R status. Furthermore, D-Allose decreased MCP-1, TNF-α, IL-1β and IL-6 levels in skin flap injured by I/R. In addition, D-Allose inhibited MKP-1 expression and activated ERK1/2 pathway in skin flap injured by I/R. PD-98059 partially counteracted D-Allose effects on I/R injury. Conclusions: D-Allose exerted its protective function via inhibiting MKP-1expression and further activated ERK1/2 pathway to suppress the progress of oxidative stress, inflammation and necrosis, contributing to the survival of skin flap injured by I/R. Thus, D-Allose was promising in the transplantation of skin flap.

Production of D-Allose From D-Allulose Using Commercial Immobilized Glucose Isomerase

Front Bioeng Biotechnol 2021 Jul 15;9:681253.PMID:34336800DOI:10.3389/fbioe.2021.681253.

Rare sugars are regarded as functional biological materials due to their potential applications as low-calorie sweeteners, antioxidants, nucleoside analogs, and immunosuppressants. D-Allose is a rare sugar that has attracted substantial attention in recent years, owing to its pharmaceutical activities, but it is still not widely available. To address this limitation, we continuously produced D-Allose from D-allulose using a packed bed reactor with commercial glucose isomerase (Sweetzyme IT). The optimal conditions for D-Allose production were determined to be pH 8.0 and 60°C, with 500 g/L D-allulose as a substrate at a dilution rate of 0.24/h. Using these optimum conditions, the commercial glucose isomerase produced an average of 150 g/L D-Allose over 20 days, with a productivity of 36 g/L/h and a conversion yield of 30%. This is the first report of the successful continuous production of D-Allose from D-allulose by commercial glucose isomerase using a packed bed reactor, which can potentially provide a continuous production system for industrial applications of D-Allose.

Recent research on the physiological functions, applications, and biotechnological production of D-Allose

Appl Microbiol Biotechnol 2018 May;102(10):4269-4278.PMID:29577167DOI:10.1007/s00253-018-8916-6.

D-Allose is a rare monosaccharide, which rarely appears in the natural environment. D-Allose has an 80% sweetness relative to table sugar but is ultra-low calorie and non-toxic and is thus an ideal candidate to take the place of table sugar in food products. It displays unique health benefits and physiological functions in various fields, including food systems, clinical treatment, and the health care fields. However, it is difficult to produce chemically. The biotechnological production of D-Allose has become a research hotspot in recent years. Therefore, an overview of recent studies on the physiological functions, applications, and biotechnological production of D-Allose is presented. In this review, the physiological functions of D-Allose are introduced in detail. In addition, the different types of D-allose-producing enzymes are compared for their enzymatic properties and for the biotechnological production of D-Allose. To date, very little information is available on the molecular modification and food-grade expression of D-allose-producing enzymes, representing a very large research space yet to be explored.