Size | Price | Stock | Qty |
---|---|---|---|
10mg |
|
||
25mg |
|
||
50mg |
|
||
100mg |
|
||
250mg |
|
||
500mg |
|
||
1g |
|
||
Other Sizes |
|
Purity: ≥98%
Myricitrin (Myricitroside), a naturally occurring flavonoid compound isolated from the root bark of Myrica cerifera, is a bioactive compound which exerts antinociceptive effects. Myricitrin produces pronounced antinociception against chemical and mechanical models of pain in rodents via preventing the protein kinase C (PKC) alpha and PKC epsilon activation by phorbol myristate acetate (PMA). Another study proves that opening of voltage- and small-conductance calcium-gated K(+) channels and reduction of calcium influx leads to the antinociceptive of myricitrin.
Targets |
Natural occurring flavonoid; Nitric oxide (NO); PKC
|
||
---|---|---|---|
ln Vitro |
In vitro activity: Myricitrin, a flavonoid compound isolated from the root bark of Myrica cerifera, which exerts antinociceptive effect. Myricitrin produces pronounced antinociception against chemical and mechanical models of pain in rodents via preventing the protein kinase C (PKC) alpha and PKC epsilon activation by phorbol myristate acetate (PMA). Another study proves that opening of voltage- and small-conductance calcium-gated K(+) channels and reduction of calcium influx leads to the antinociceptive of myricitrin. Myricitrin also decreases H2O2-induced apoptosis in vascular endothelial cells via inhibition of p53 gene expression, activation of caspase-3 and the MAPK signaling pathway, and alteration of the patterns of pro-apoptotic and anti-apoptotic gene expression.
Myr/Myricitrin treatment inhibited the lamellipodia formation, migration, and invasion, but not the apoptosis and proliferation, of RA FLSs. Myr also reduced the expression of CCL2, IL-6, IL-8, MMP-1, MMP-3, and MMP-13 induced by TNF-α. The RNA-seq results indicated that AIM2 may be a target gene of Myr in RA FLSs. Furthermore, compared to healthy controls, AIM2 expression showed higher levels in synovial tissues and FLSs from RA patients. AIM2 knockdown also inhibited RA FLS migration, invasion, cytokine, and MMP expression. In addition, either Myr treatment or AIM2 knockdown reduced the phosphorylation of AKT induced by TNF-α stimulation [3]. |
||
ln Vivo |
In mice, Myricitrin (i.p. 10 mg/kg or 30 mg/kg) blocks apomorphine-induced stereotypy and climbing, and increases hindlimb retraction time (HRT).
The present study investigated the antinociceptive effects of the flavonoid myricitrin in chemical behavioral models of pain in mice and rats. Myricitrin given by i.p. or p.o. routes produced dose-related antinociception when assessed on acetic acid-induced visceral pain in mice. In addition, the i.p. administration of Myricitrin exhibited significant inhibition of the neurogenic pain induced by intraplantar (i.pl.) injection of capsaicin. Like-wise, myricitrin given by i.p. route reduced the nociception produced by i.pl. injection of glutamate and phorbol myristate acetate (PMA). Western blot analysis revealed that myricitrin treatment fully prevented the protein kinase C (PKC) alpha and PKCepsilon activation by PMA in mice hind paws. Myricitrin given i.p. also inhibited the mechanical hyperalgesia induced by bradykinin, without affecting similar responses caused by epinephrine and prostaglandin E(2). The antinociception caused by myricitrin in the acetic acid test was significantly attenuated by i.p. treatment of mice with the nitric oxide precursor, L-arginine. In contrast, myricitrin antinociception was not affected by naloxone (opioid receptor antagonist) or neonatal pretreatment of mice with capsaicin and myricitrin antinociceptive effects is not related to muscle relaxant or sedative action. Together, these results indicate that myricitrin produces pronounced antinociception against chemical and mechanical models of pain in rodents. The mechanisms involved in their actions are not completely understood but seem to involve an interaction with nitric oxide-L-arginine and protein kinase C pathways. [1] The present study was designed to investigate the mechanisms involved in the antinociception afforded by Myricitrin in chemical models of nociception in mice. Myricitrin given by intrathecal (i.t.) or intracerebroventricular (i.c.v.) route produced dose-related antinociception when evaluated against acetic acid-induced visceral pain in mice. In addition, the intraperitoneal administration of myricitrin caused significant inhibition of biting behaviour induced by i.t. injection of glutamate, substance P, capsaicin, interleukin 1 beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha). The antinociception caused by myricitrin in the acetic acid test was fully prevented by i.t. pre-treatment with pertussis toxin, a Gi/o protein inactivator, and by i.c.v. injection of calcium chloride (CaCl(2)). In addition, the i.t. pre-treatment of mice with apamin, a blocker of small (or low)-conductance calcium-gated K(+) channels and tetraethylammonium, a blocker of voltage-gated K(+) channels significantly reversed the antinociception induced by myricitrin. The charybdotoxin, a blocker of large (or fast)-conductance calcium-gated K(+) channels and glibenclamide, a blocker of the ATP-gated K(+) channels had no effect on myricitrin-induced antinociception. Calcium uptake analysis revealed that myricitrin inhibited (45)Ca(2+) influx under a K(+)-induced depolarization condition. However, calcium movement was modified in a non-depolarizing condition only when the highest concentration of myricitrin was used. In summary, our findings indicate that myricitrin produces consistent antinociception in chemical models of nociception in mice. These results clearly demonstrate an involvement of the Gi/o protein dependent mechanism on antinociception caused by myricitrin. The opening of voltage- and small-conductance calcium-gated K(+) channels and the reduction of calcium influx led to the antinociceptive of myricitrin [2]. |
||
Cell Assay |
RA FLS viability assay [3]
Cell Counting Kit-8 (CCK8) assay kit was used to assess RA FLS viability. Briefly, RA FLSs were incubated with Myricitrin/Myr at different concentrations (0–800 μM) for 24 h. Cells were washed with DMEM. Subsequently, a culture medium containing 10% Cell Counting Kit-8 (CCK-8) reagent was added to each well and incubated for another 4 h. Afterward, the absorbance of the plate was measured at 450 nm by a microplate reader. FLS migration and invasion assay [3] FLS migration was performed using a transwell assay with a filter (6.5 mm in diameter, 8.0 μm pore size). Briefly, FLSs were suspended in serum-free DMEM in the upper compartments of the chambers at a concentration of 6×104 cells/ml. Medium containing 10% FBS as a chemoattractant was added to the lower compartments of the chambers. After incubation for 8 h, the cells on the top surface of the membrane were scraped using a cotton swab. RA FLSs that migrated to the lower side of the filter were fixed in methanol for 15 min and stained with 0.1% crystal violet for 15 min. The number of stained FLSs is the average number of cells from 5 random fields. To measure cell invasion, we performed a similar experiment in chambers coated with BD Matrigel basement membrane matrix. FLS proliferation assays [3] RA FLS proliferation was measured by using a Cell-Light EdU DNA Cell Proliferation Kit following the manufacturer’s protocol. The cells were pretreated with different concentrations of Myricitrin/Myr (0–400 μM) for 24 h. EdU was added to measure the cell proliferation and incubated for another 12 h. DAPI was used to stain cell nuclei. EdU-positive cells were quantified by microscopy. FLS apoptosis assays [3] An Annexin V-APC/PI Apoptosis Detection kit was used to assess RA FLS apoptosis following the manufacturer’s protocol. Briefly, 1 × 105 FLSs were suspended in 0.1 ml 1× binding buffer, stained with 5 μL PI and 5 μL annexin V and incubated for another 15 min in darkness at room temperature. The samples were then analyzed within 1 h by flow cytometry. |
||
Animal Protocol |
|
||
References |
[1]. J Pharmacol Exp Ther.2006 Feb;316(2):789-96.
[2]. Eur J Pharmacol.2007 Jul 19;567(3):198-205. [3]. Front Pharmacol. 2022 Aug 31:13:905376. |
||
Additional Infomation |
Myricitrin is a glycosyloxyflavone that consists of Myricitrin attached to a alpha-L-rhamnopyranosyl residue at position 3 via a glycosidic linkage. Isolated from Myrica cerifera, it exhibits anti-allergic activity. It has a role as an anti-allergic agent, an EC 1.14.13.39 (nitric oxide synthase) inhibitor, an EC 2.7.11.13 (protein kinase C) inhibitor and a plant metabolite. It is a pentahydroxyflavone, a glycosyloxyflavone, an alpha-L-rhamnoside and a monosaccharide derivative. It is functionally related to a myricetin. It is a conjugate acid of a myricitrin(1-).
Myricitrin has been reported in Castanopsis fissa, Phyllanthus tenellus, and other organisms with data available. In conclusion, the present results are in agreement with previous data and demonstrate that Myricitrin produces antinociception when administered at peripheral or central levels. Furthermore, these results showed that myricitrin antinociception is closely related to pathways activated by glutamate, substance P, capsaicin and pro-inflammatory cytokines. The mechanisms of antinociception are dependent of the Gi/o protein activation; opening of specific K+ channels (voltage- and small-conductance Ca2+-gated) and inhibition of calcium influx.[2] Objective: To explore the effect and underlying mechanism of Myricitrin (Myr) in regulating fibroblast-like synoviocyte (FLS)-mediated synovitis and joint destruction in RA. Methods: FLSs were isolated from synovial tissues from patients with RA. Gene expression was measured using quantitative RT-qPCR. Protein expression was detected by immunohistochemistry or Western blot. Cell apoptosis was performed by an Annexin-PI staining assay. EdU incorporation was used to assess the proliferation of RA FLS. Transwell assay was used to characterize the cell migration and invasion ability of RA FLS. The potential target of Myr was identified by RNA sequencing analysis. The in vivo effect of Myr was assessed in a collagen-induced arthritis (CIA) model. Results: Myr treatment inhibited the lamellipodia formation, migration, and invasion, but not the apoptosis and proliferation, of RA FLSs. Myr also reduced the expression of CCL2, IL-6, IL-8, MMP-1, MMP-3, and MMP-13 induced by TNF-α. The RNA-seq results indicated that AIM2 may be a target gene of Myr in RA FLSs. Furthermore, compared to healthy controls, AIM2 expression showed higher levels in synovial tissues and FLSs from RA patients. AIM2 knockdown also inhibited RA FLS migration, invasion, cytokine, and MMP expression. In addition, either Myr treatment or AIM2 knockdown reduced the phosphorylation of AKT induced by TNF-α stimulation. Importantly, Myr administration relieved arthritis symptoms and inhibited AIM2 expression in the synovium of CIA mice. Conclusion: Our results indicate that Myr exerts an anti-inflammatory and anti-invasion effect in RA FLSs and provide evidence of the therapeutic potential of Myr for RA.[3] |
Molecular Formula |
C21H20O12
|
|
---|---|---|
Molecular Weight |
464.38
|
|
Exact Mass |
464.095
|
|
Elemental Analysis |
C, 54.32; H, 4.34; O, 41.34
|
|
CAS # |
17912-87-7
|
|
Related CAS # |
|
|
PubChem CID |
5281673
|
|
Appearance |
White to yellow solid powder
|
|
Density |
1.9±0.1 g/cm3
|
|
Boiling Point |
896.6±65.0 °C at 760 mmHg
|
|
Melting Point |
197 °C
|
|
Flash Point |
315.7±27.8 °C
|
|
Vapour Pressure |
0.0±0.3 mmHg at 25°C
|
|
Index of Refraction |
1.805
|
|
LogP |
1.98
|
|
Hydrogen Bond Donor Count |
8
|
|
Hydrogen Bond Acceptor Count |
12
|
|
Rotatable Bond Count |
3
|
|
Heavy Atom Count |
33
|
|
Complexity |
760
|
|
Defined Atom Stereocenter Count |
5
|
|
SMILES |
C[C@H]1[C@@H]([C@H]([C@H]([C@@H](O1)OC2=C(OC3=CC(=CC(=C3C2=O)O)O)C4=CC(=C(C(=C4)O)O)O)O)O)O
|
|
InChi Key |
DCYOADKBABEMIQ-OWMUPTOHSA-N
|
|
InChi Code |
InChI=1S/C21H20O12/c1-6-14(26)17(29)18(30)21(31-6)33-20-16(28)13-9(23)4-8(22)5-12(13)32-19(20)7-2-10(24)15(27)11(25)3-7/h2-6,14,17-18,21-27,29-30H,1H3/t6-,14-,17+,18+,21-/m0/s1
|
|
Chemical Name |
3-[(6-deoxy-α-L-mannopyranosyl)oxy]-5,7-dihydroxy-2-(3,4,5-trihydroxyphenyl)-4H-1-benzopyran-4-one
|
|
Synonyms |
|
|
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 Note: This product requires protection from light (avoid light exposure) during transportation and storage. |
|
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) |
|
|||
---|---|---|---|---|
Solubility (In Vivo) |
Solubility in Formulation 1: ≥ 2.5 mg/mL (5.38 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.  (Please use freshly prepared in vivo formulations for optimal results.) |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 2.1534 mL | 10.7670 mL | 21.5341 mL | |
5 mM | 0.4307 mL | 2.1534 mL | 4.3068 mL | |
10 mM | 0.2153 mL | 1.0767 mL | 2.1534 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.