Size | Price | Stock | Qty |
---|---|---|---|
25mg |
|
||
50mg |
|
||
100mg |
|
||
250mg |
|
||
500mg |
|
||
Other Sizes |
|
Targets |
NF-κB
|
---|---|
ln Vitro |
Barlerin (8-O-Acetyl shanzhiside methyl ester) treatment inhibits TNF-induced nuclear transcription factor B (NF-κB) activation and reduces high-mobility group box-1 (HMGB-1) expression in SH-SY5Y cells. [1]. By preventing High-mobility group box 1 (HMGB1) expression, treatment of H9c2 cells with barlerin (8-O-Acetyl shanzhiside methyl ester) 9 μM prevents TNF-α-induced NF-κB phosphorylation[2].
Effects of 8-O-acetyl shanzhiside methylester (ND01) on NF-κB activation and HMGB-1 expression [1] The NF-κB pathway plays a critical role in the secretion of cytokines. The quantity of p50 and p65 was measured in the nucleus. Stimulation with TNF-α led to a robust activation of the NF-κB transcription factor p50/p65. However, this activation was partially blocked by ND01, as shown in fig. 1. [1] The IκB kinase system as another activation agent of NF-κB was also examined. Stimulation with TNF-α resulted in a marked degradation of IκB. This degradation was inhibited by ND01 (fig. 2). In addition, the phosphorylation of p-IκB was increased by TNF-α and also inhibited by ND01. [1] Phosphor-NF-κB expression was low in non-TNF-α-stimulated SH-SY5Y cells. However, phosphor-NF-κB expression was significantly increased after TNF-α 20 ng/ml-stimulated SH-SY5Y cells for 120 min. We compared the effect of ND01 on the TNF-α-induced activation of phosphor-NF-κB and HMGB-1 expression level in the SH-SY5Y cells to that of a selective HMGB-1 inhibitor, glycyrrhizin. The results of fig. 2 showed that pre-treatment of SH-SY5Y cells with glycyrrhizin (100 μM) for 120 min., blocked the TNF-α-induced HMGB-1 expression and reduced phosphorylation of NF-κB. Pre-treatment of SH-SY5Y cells with ND01 10 μM blocked TNF-α-induced NF-κB phosphorylation and reduced HMGB-1 expression, as shown in fig. 2. [1] |
ln Vivo |
Barlerin (8-O-Acetyl shanzhiside methyl ester) 40 mg/kg exhibits a notable neuroprotective effect even when administered 4 hours after I/R. In ischaemic brain tissue, barlerin 40 mg/kg inhibits NF-κB activation, reduces HMGB-1 expression, attenuates histopathological damage, and reduces brain swelling[1]. The angiogenesis in the ischaemic brain is markedly accelerated by barlerin (8-O-Acetyl shanzhiside methyl ester), which also enhances functional recovery after stroke. Additionally, compared to vehicle treatment, barlerin significantly boosts vascularization. It raises the levels of Ang1, Tie2, and Akt VEGF as well as their expression[3]. Activated partial thromboplastin, prothrombin, and thrombin times in mice are unaffected by barlerin (8-O-Acetyl shanzhiside methyl ester), but capillary blood clotting time and blood loss volume are significantly shortened. In hyperfibrinolysis mice, it significantly extends the time it takes for euglobulin clots to dissolve[4].
Inflammatory activation plays a vital role in the pathophysiological mechanisms of stroke and diabetes mellitus (DM), exerts the deleterious effects on the progression of the brain and leads to vascular damage in diabetic stroke. The objectives of this study were to investigate the effects of 8-O-acetyl shanzhiside methylester (ND01) on tumour necrosis factor-α (TNF-α)-stimulated SH-SY5Y cell line in vitro and the experimental ischaemic diabetic stroke model in vivo. TNF-α-stimulated SH-SY5Y cells were pre-incubated with ND01, then analysed protein expression. For in vivo experiment, the diabetic rats were subjected to middle cerebral artery occlusion (MCAO) for 30 min. followed by reperfusion for 23 hr. Treatment of SH-SY5Y cells with ND01 blocked TNF-α-induced nuclear transcription factor κB (NF-κB) activation and decreased high-mobility group box-1 (HMGB-1) expression. ND01 40 mg/kg demonstrated significant neuroprotective effect even after delayed administration at 4 hr after I/R. ND01 40 mg/kg attenuated the histopathological damage, decreased brain swelling, inhibited NF-κB activation and reduced HMGB-1 expression in ischaemic brain tissue. These data show that ND01 protects diabetic brain against I/R injury with a favourable therapeutic time-window by alleviating diabetic cerebral I/R injury and attenuating blood-brain barrier (BBB) breakdown, and its protective effects may involve HMGB-1 and NF-κB signalling pathway.[1] 8-O-acetyl shanzhiside methylester (ND01) was isolated from the leaves of Lamiophlomis rotata (Benth.) Kudo. In this study, we investigated the anti-myocardial ischemia and reperfusion (I/R) injury effects of ND01 in vivo and elucidated the potential mechanism in vitro. The results indicated that ND01 significantly attenuated hypoxia-induced cytotoxicity in a concentration-dependent manner in H9c2 cells. Treatment of H9c2 cells with ND01 9 μM blocked TNF-α-induced nuclear factor kappaB (NF-κB) phosphorylation by blocking High-mobility group box1 (HMGB1) expression. Treatment of rats with ND01 10mg/kg, (i.v.) protected the animals from myocardial I/R injury as indicated by a decrease in infarct volume, improvement in hemodynamics and reduction of myocardial damage severity. Treatment with ND01 also lowered serum levels of pro-inflammatory factors and reduced High mobility group box-1 protein (HMGB1) and phosphorylated NF-κB expression in ischemic myocardial tissue. Additionally, continuous i.v. of ND01 14 days attenuated cardiac remodeling. These protective effects suggested that ND01 might be due to block of myocardial inflammatory cascades through an HMGB1-dependent NF-κB signaling pathway.[2] Reseasrchers investigated whether 8-O-acetyl shanzhiside methylester (ND01) regulates angiogenesis and thereby improves functional outcome after stroke. Adult male rats were subjected to 1 hr of middle cerebral artery occlusion (MCAO) and reperfusion, and treated with or without different doses (5 and 10 mg/kg) of ND01, starting 24 hr after ischaemia and reperfusion (I/R) and by intravenous injection daily for 14 days. Neurological functional tests were performed and cerebral Evans blue extravasation was measured. Angiogenesis and angiogenic factor expression were measured by immunohistochemistry and Western blot, respectively. The results indicated that ND01 significantly promoted angiogenesis in the ischaemic brain and improved functional outcome after stroke. ND01 also significantly increased vascularization compared with vehicle treatment. ND01 increased the expression of VEGF, Ang1, phosphorylation of Tie2 and Akt VEGF. The Ang1/Tie2 axis and Akt pathways appear to mediate ND01-induced angiogenesis. [3] The anti-fibrinolytic activity come from 8-O-Acetyl shanzhiside methylester (ASM) one of the highest iridoid glycosides contents in TIG extracted from L. rotata. ASM significantly (P<0.05) shorten CBCT and reduced blood loss volume in vivo, but did not influence mice APTT, PT or TT. In particular, it significantly prolonged ECLT in hyperfibrinolysis mice. It indicated that ASM could inhibit fibrinolysis. ASM was also effective in CBCT, traumatic bleeding volume and ECLT in hyperfibrinolysis mice model. Conclusions: ASM was the major hemostatic compound in L. rotata. The haemostasis mechanism of ASM was achieved by anti-fibrinolytic activity. ASM was a new fibrinolysis inhibitor as iridoid glycoside compound.[4] |
Enzyme Assay |
NF-κB-binding assay [1]
SH-SY5Y cells (5 × 106) were pre-incubated with 8-O-acetyl shanzhiside methylester (ND01) 10 μM for 22 hr, then incubated with TNF-α (20 ng/ml) for 1 or 2 hr, then washed once with PBS, scraped cells into 1 ml cold PBS and pelleted by centrifugation. Nuclear extracts were prepared as described previously 12. The DNA-binding activity of NF-κB (p50/p65) was determined using an ELISA kit 8-O-acetyl shanzhiside methylester (ND01) inhibited NF-κB activation and HMGB1 expression [2] We compared the effect of ND01 on the TNF-α-induced (20 ng/ml for 30 min) activation of NF-κB and HMGB1 in H9c2 cells to a selective HMGB1 inhibitor glycyrrhizin (100 μM) and a NF-κB |
Cell Assay |
Cells are pretreated with Barlerin (8-O-Acetyl shanzhiside methyl ester) in a range of concentrations (1, 3, 9 and 27 27μM) for 24 hours prior to hypoxia. MTT assays are used to assess cell viability[2].
NF-κB activation and HMGB-1 expression in TNF-α-stimulated SH-SY5Y cells [1] Human neuroblastoma (SH-SY5Y) cells were were cultured and maintained in F12+ DMEM (1:1, v/v) media, supplemented with 10% phosphate buffered saline (PBS) and 1% penicillin/streptomycin. Cells were kept at 37°C in a humidified 5% CO2/95% O2 incubator. The dissociated cells were seeded in poly-l-lysine-coated plates at a density of 5 × 105/cm2 and cultured in DMEM, supplemented with 10% (v/v) horse serum, 5% (v/v) foetal bovine serum, 100 U/ml penicillin and 0.1 mg/ml streptomycin. The fresh medium was changed twice weekly. [1] For the experiment of TNF-α-stimulated SH-SY5Y cell lines in vitro, SH-SY5Y cells (5 × 106) were pre-incubated with 8-O-acetyl shanzhiside methylester (ND01) (9 μM) or HMGB-1 inhibitor, glycyrrhizin (100 μM) for 120 min., and then incubated with TNF-α (20 ng/ml) for 30 min. and cultured in a CO2 incubator for 12 hr. Cells were washed twice with ice-cold PBS on ice and lysed in NP40 lysis buffer 50 mM Tris, pH 7.4, 250 mM NaCl, 5 mM EDTA, 50 mM NaF, 1 mM Na3VO4, 1% NP-40 and 0.02% NaN3) supplemented with 1 mM PMSF and 1 × protease inhibitor cocktail. Equal amounts of cell protein (50 μg) were separated by SDS–PAGE and analysed by Western blot using specific antibodies to HMGB-1, IκB, phosphor-IκB-α, phosphor-NF-κB and proliferating cell nuclear antigen (PCNA, loading control). Optical densities of the bands were scanned and quantified with a Gel Doc 2000. Data were normalized against those of the corresponding PCNA bands. Results were expressed as fold increase over control. 8-O-acetyl shanzhiside methylester (ND01) attenuated hypoxia-induced cytotoxicity [2] The results of cell viability were shown in Fig.1. Exposed to hypoxia for 6 h, there were only 52.0 ± 8.9% viable cells as compared to the control cells. 8-O-acetyl shanzhiside methylester (ND01) (3, 9 and 27 μM) prevented cells from hypoxia-induced damage in a concentration-dependent manner, restored cell survival to 63.7 ± 6.0%, 69.9 ± 8.6% and 74.4 ± 9.6%. |
Animal Protocol |
Rats: In saline, Barlerin (8-O-Acetyl shanzhiside methyl ester) is made. Adult male rats are given an hour of middle cerebral artery occlusion (MCAO) and reperfusion. They are then given different doses of 8-O-acetyl shanzhiside methylester (ND01) (5 and 10 mg/kg) intravenously every day for 14 days, starting 24 hours after the I/R (ischaemia and reperfusion). In addition to measuring cerebral Evans blue extravasation, neurological functional tests are carried out[3].
Mouse: In saline, Barlerin (8-O-Acetyl shanzhiside methyl ester) is made. The five groups (saline group, Hemocoagulase, 0.34 KU/kg, intravenous, ASM-L, 100 mg/kg, ASM-M, 250 mg/kg, and ASM-H, 500 mg/kg) of male Balb/C mice (20 to 25g) are assigned at random. Five minutes before sodium pentobarbital (200 mg/kg, i.p.) is used to anesthetize the patient, the drugs and the vehicle are injected through the vena cauda. After the injection, blood is drawn from the heart 20 minutes later. Prothrombin time, thrombin time, fibrinogen, and activated partial thromboplastin time are measured[4]. Streptozotocin (STZ)-induced diabetes model and rat cerebral ischaemia study protocol [1] Two hundred rats (fasting for 20 hr) were induced by single i.p. STZ at a dose of 50 mg/kg. STZ was diluted in citrate buffer 0.1 M (pH 4.0). After STZ injection for 3 weeks, rats with glycaemia value between 12.0 and 20.0 mM were used. The diabetic rats were anaesthetized with chloral hydrate (350 mg/kg, i.p.). Rectal temperature was recorded and maintained at 37°C throughout the surgical procedure. The operation of MCAO was carried out according to previous procedures with minor modifications 13. The left common carotid artery was occluded, and the branches of the external carotid artery were dissected and divided. The internal carotid artery was followed rostrally, and a 4-0 filament (the diameter of the filament was 0.25, but the diameter of the tip was 0.34 mm to create a globular stopper) was introduced into the internal carotid artery and advanced until resistance was felt. The filament was removed after 30 min. The rats were kept under conditions of controlled temperature (24–25°C) for the first 23 hr after surgery. [1] A pilot study was conducted with four different doses of 8-O-acetyl shanzhiside methylester (ND01) (10, 20, 40 or 80 mg/kg) to determine the dose-dependent effect in the acute I/R-treated diabetic rats. It was observed that 8-O-acetyl shanzhiside methylester (ND01) at doses of 20, 40 and 80 mg/kg significantly (p < 0.05) lowered infarct volume and neurological deficit scores of the acute I/R-induced diabetic rats after 23 hr of the experiment. Hence, 8-O-acetyl shanzhiside methylester (ND01) 40 mg/kg was chosen for this study. [1] For therapeutic time-window studies, 40 MCAO diabetic rats were randomly divided into four groups of 10 rats each plus 10 diabetic rats as control. Rats received dose of 40 mg/kg by intravenous bolus injection into the tail vein 2, 4 and 6 hr after reperfusion. Vehicle-treated rats were administered with saline. Neurological deficits were determined 23 hr after ischaemia followed by brain infarct examination. [1] For anti-inflammatory mechanism studies, 54 rats were randomly divided into three subgroups of 18 rats each plus 18 rats as control (non-diabetic). Rats received a dose of 40 mg/kg intravenous bolus injection into the tail vein 30 min. after reperfusion. Diabetic or vehicle-treated rats were administered with saline. All the brain were evaluated by Evans blue extravasation, then analysed HMGB1, IκB, phosphor-IκB-α, phosphor-NF-κB by Western blot and the histopathological damage were evaluated by NeuN staining, especially. [1] For long-term studies, 20 rats were randomly divided into two groups of 10 rats each. Rats received a dose of 40 mg/kg by intravenous bolus injection into the tail vein 30 min. after reperfusion. The vehicle-treated rats were administered with saline. Neurological deficits were determined on days 3, 7 and 14 after I/R. Fourteen days after I/R, eight rats were stained in the 8-O-acetyl shanzhiside methylester (ND01) 40 mg/kg group, and six rats were stained in the vehicle-treated group. The brain infarct was examined according to a previous method. [1] I/R procedure to induce cerebral ischaemia. [3] The body weight of rats was 280–320 g. After 1 week of acclimatization, rats were anaesthetized with chloral hydrate (350 mg/kg, i.p.). The middle cerebral artery occlusion (MCAO) operation was performed according to procedures described previously. The left common carotid artery was occluded, and the branches of the external carotid artery were dissected and divided. The internal carotid artery was followed rostrally and a 4–0 filament (the diameter of the filament is 0.25, but the diameter of the tip is 0.34 mm to create a globular stopper) was introduced into the internal carotid artery and advanced until resistance was felt. The filament was removed after 1 hr. Core body temperature was maintained at 37 ± 0.5°C on the heating pad. A total of 160 rats were divided into two groups; each group included four subgroups (each subgroup consisted of 20 rats): non-I/R (sham) group, vehicle-treated group, 8-O-acetyl shanzhiside methylester (ND01) 5 mg/kg group and 8-O-acetyl shanzhiside methylester (ND01) 10 mg/kg group. After 23 hr of reperfusion, all animals were administered an intravenous bolus injection (i.v., via the tail) of the corresponding drug daily for 14 days. The sham and vehicle-treated rats were both given saline. At 7 and 14 days, the first group of animals was used to evaluate nerve behaviour, then to measure permeability of the blood-brain barrier; each subgroup consisted of eight rats. At 7 and 14 days, the second group of animals was used to analyse Western blots, microvasculature density and immunohistochemical staining. Four rats of each subgroup were used to analyse Western blot in ischaemic brain core; the next four rats of each subgroup were used to detect microvasculature density and immunohistochemical staining.[3] |
References |
|
Additional Infomation |
8-O-Acetyl shanzhiside methyl ester has been reported in Barleria lupulina, Lamium garganicum, and other organisms with data available.
In the present study, we observed that 8-O-acetyl shanzhiside methylester (ND01) significantly improved brain injury and did not reduce blood glucose in diabetic rats subjected to cerebral I/R challenge, indicating that ND01 has an immediate neuroprotective effect but not by reducing blood glucose. Infarct size is the important indicator of the pathophysiology to evaluate the efficacy of cerebral ischaemia 19. Our results (fig. 3) revealed that 8-O-acetyl shanzhiside methylester (ND01) treatment produced significant reduction in the cerebral infarct volume in diabetic cerebral I/R rats even delayed administration at 2 and 4 hr after I/R. Neuronal degeneration and necrosis have been found to be correlated with deficits in behavioural disturbance, and behavioural assessment may reveal effectiveness 20. The present study showed that neurological scores were reduced by treatment with ND01 (fig. 3). NeuN is a sensitive marker for injured neuron early after ischaemic challenge 21, and our data (fig. 4) demonstrate that ND01 at a dose of 40 mg/kg attenuated the decrease in NeuN-immunopositive neurones in ischaemic cerebral cortex at 23 hr and 14 days after I/R. This indicates that ND01 has potential beneficial effects in the treatment of cerebral ischaemia. [1] The update of the Stroke Therapy Academic Industry Roundtable Preclinical Recommendations 22 lists potential reasons for failures in translating efficacious preclinical findings into successful clinical trial outcome. Consequently, we investigate the therapeutic time-window and the long-term efficacy of 8-O-acetyl shanzhiside methylester (ND01) in the cerebral I/R diabetic rats, and our data demonstrate that 8-O-acetyl shanzhiside methylester (ND01) exerted potent and long-term neuroprotective effects with a favourable therapeutic time-window. [1] Stroke triggers an inflammatory reaction that progresses for hours after the onset of the stroke, and this inflammation plays a central role in the pathogenesis of neuronal injury in ischaemic stroke, especially in diabetic stroke. Inflammatory reactions contribute to the late stages of ischaemic injury and worsened neurological outcome through the multiple mechanisms. DM is also an inflammatory disease. The present study indicates that 8-O-acetyl shanzhiside methylester (ND01) had significant anti-inflammatory effects (table 2 and fig. 5), and treatment with ND01 especially provides long-term benefits for neuronal functional recovery after cerebral I/R (table 1). This suggests that the neuroprotective effects of ND01 might be due to blocking of the inflammatory response. [1] MPO is considered an index of neutrophil infiltration and highly expressed cerebral ischaemia at 24 hr. It shows that a significant correlation between neutrophil infiltration and infarct formation exists in a model of cerebral ischaemia. Our results show that ND01 reduced MPO activity in diabetic ischaemic cerebral tissue. This suggests that the neuroprotective effects of ND01 might be due to blocking of neutrophil infiltration (table 2). [1] BBB permeability is significantly increased in diabetes due to diabetes-induced damage to the BBB function and/or due to the immature nature of the newly formed vessel. BBB breakdown also occurs in early-phase (within 24 hr) of cerebral ischaemia. Our data demonstrate that ND01 improved diabetic cerebral I/R injury by attenuating BBB breakdown. [1] NF-κB activation is associated with the phosphorylation of IκB-α and NF-κB in ischaemic cerebral tissue. Reduction in NF-κB activation can protect brain from activation of NF-κB-dependent genes. HMGB-1 is a novel player in the ischaemic brain. Meanwhile, diabetes significantly increased HMGB levels and induced worse functional outcome after stroke compared with non-diabetic MCAO rats. All those data show HMGB-1 plays a key role in diabetic stroke. The HMGB-1 signalling involves the activation of NF-κB, an inhibitor of NF-κB kinase essential for HMGB-1. Suppression the release of HMGB-1 in astrocytes leads to the attenuation of neuroinflammation and prevents the necrosis of ischaemic astrocytes and NF-κB expression 34. Inhibition of the up-regulation of HMGB-1 and NF-κB at the early stage brings great benefits to the diabetic cerebral ischaemia. Based on the above findings, we explored the anti-inflammatory properties of 8-O-acetyl shanzhiside methylester (ND01) in diabetic cerebral ischaemia and further studied the potential mechanisms. The up-regulation of HMGB-1 and NF-κB is significantly suppressed by 8-O-acetyl shanzhiside methylester (ND01). These results suggest that suppressing HMGB-1 and NF-κB expressions participated in the neuroprotection of ND01 against diabetic cerebral ischaemic damage. Therefore, we believe that the protective effects of ND01 might be due to suppression of the inflammatory cascades through HMGB-1-dependent NF-κB signalling pathway. [1] In summary, the results of the current study suggest that 8-O-acetyl shanzhiside methylester (ND01) exhibits significant neuroprotective effects during diabetic cerebral I/R injury, including attenuation of BBB breakdown, decrease in the infarct volume, alleviation of cerebral damage, reduction in HMGB-1 expression, phosphorylated IκB-α and NF-κB expression in ischaemic brain tissue. These effects of ND01 are correlated with inhibition of the inflammatory response. These findings point to a therapeutic potential for ND01 as a useful anti-inflammatory leads compound in early diabetic cerebral I/R injury. |
Molecular Formula |
C₁₉H₂₈O₁₂
|
---|---|
Molecular Weight |
448.42
|
Exact Mass |
448.158
|
CAS # |
57420-46-9
|
Related CAS # |
57420-46-9
|
PubChem CID |
162823
|
Appearance |
White to off-white solid
|
Density |
1.52 g/cm3
|
Boiling Point |
634.2±55.0 °C at 760 mmHg
|
Flash Point |
220.0±25.0 °C
|
Vapour Pressure |
0.0±4.2 mmHg at 25°C
|
Index of Refraction |
1.594
|
LogP |
-2.76
|
Hydrogen Bond Donor Count |
5
|
Hydrogen Bond Acceptor Count |
12
|
Rotatable Bond Count |
7
|
Heavy Atom Count |
31
|
Complexity |
725
|
Defined Atom Stereocenter Count |
10
|
SMILES |
O(C(C([H])([H])[H])=O)[C@@]1(C([H])([H])[H])C([H])([H])[C@]([H])([C@]2([H])C(C(=O)OC([H])([H])[H])=C([H])O[C@]([H])([C@]12[H])O[C@@]1([H])[C@@]([H])([C@]([H])([C@@]([H])([C@@]([H])(C([H])([H])O[H])O1)O[H])O[H])O[H])O[H]
|
InChi Key |
ARFRZOLTIRQFCI-NGQYDJQZSA-N
|
InChi Code |
InChI=1S/C19H28O12/c1-7(21)31-19(2)4-9(22)11-8(16(26)27-3)6-28-17(12(11)19)30-18-15(25)14(24)13(23)10(5-20)29-18/h6,9-15,17-18,20,22-25H,4-5H2,1-3H3/t9-,10-,11+,12-,13-,14+,15-,17+,18+,19+/m1/s1
|
Chemical Name |
methyl (1S,4aS,5R,7S,7aS)-7-acetyloxy-5-hydroxy-7-methyl-1-[(2S,3R,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-4a,5,6,7a-tetrahydro-1H-cyclopenta[c]pyran-4-carboxylate
|
Synonyms |
8-O-Acetyl shanzhiside methyl ester; 8-O-Acetyl shanzhiside methyl ester; 8-O-Acetylshanzhiside methyl ester; methyl (1S,4aS,5R,7S,7aS)-7-acetyloxy-5-hydroxy-7-methyl-1-[(2S,3R,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-4a,5,6,7a-tetrahydro-1H-cyclopenta[c]pyran-4-carboxylate; 5beta-Dihydro Finasteride; Cyclopenta[c]pyran-4-carboxylic acid,7-(acetyloxy)-1-(b-D-glucopyranosyloxy)-1,4a,5,6,7,7a-hexahydro-5-hydroxy-7-methyl-, methyl ester, (1S,4aS,5R,7S,7aS)-; Umbroside; Barlerin
|
HS Tariff Code |
2934.99.9001
|
Storage |
Powder -20°C 3 years 4°C 2 years In solvent -80°C 6 months -20°C 1 month |
Shipping Condition |
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
|
Solubility (In Vitro) |
DMSO: ~100 mg/mL (~223.0 mM)
|
---|---|
Solubility (In Vivo) |
Solubility in Formulation 1: ≥ 2.5 mg/mL (5.58 mM) (saturation unknown) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% Saline (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 400 μL PEG300 and mix evenly; then add 50 μL Tween-80 to the above solution and mix evenly; then add 450 μL normal saline to adjust the volume to 1 mL. Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution. Solubility in Formulation 2: ≥ 2.5 mg/mL (5.58 mM) (saturation unknown) in 10% DMSO + 90% (20% SBE-β-CD in Saline) (add these co-solvents sequentially from left to right, and one by one), clear solution. For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 900 μL of 20% SBE-β-CD physiological saline solution and mix evenly. Preparation of 20% SBE-β-CD in Saline (4°C,1 week): Dissolve 2 g SBE-β-CD in 10 mL saline to obtain a clear solution. View More
Solubility in Formulation 3: ≥ 2.5 mg/mL (5.58 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution. |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 2.2301 mL | 11.1503 mL | 22.3005 mL | |
5 mM | 0.4460 mL | 2.2301 mL | 4.4601 mL | |
10 mM | 0.2230 mL | 1.1150 mL | 2.2301 mL |
*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.
Calculation results
Working concentration: mg/mL;
Method for preparing DMSO stock solution: mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.
Method for preparing in vivo formulation::Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.
(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
(2) Be sure to add the solvent(s) in order.