yingweiwo

Glycochenodeoxycholic acid sodium salt

Alias: 16564-43-5; Glycochenodeoxycholic acid sodium salt; Sodium glycochenodeoxycholate; Sodium glycylchenodeoxycholate; NSC 681056; CHENYLGLYCINE SODIUM; Glycochenodeoxycholic acid (sodium salt); OK5NH65A9B;
Cat No.:V32387 Purity: ≥98%
Glycochenodeoxycholic acid sodium, the sodium salt of Glycochenodeoxycholic acid which is a glycine conjugate of lithocholic acid, is a bile salt produced in the liver from chenodeoxycholate and glycine.
Glycochenodeoxycholic acid sodium salt
Glycochenodeoxycholic acid sodium salt Chemical Structure CAS No.: 16564-43-5
Product category: Endogenous Metabolite
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
5g
10g
25g
50g
Other Sizes

Other Forms of Glycochenodeoxycholic acid sodium salt:

  • Glycochenodeoxycholic acid-d7 sodium (Chenodeoxycholylglycine-d7 (sodium); Sodium glycochenodeoxycholate-d7)
  • Glycochenodeoxycholic acid 3-sulfate-d5 disodium
  • Glycochenodeoxycholic acid-d4 (Chenodeoxycholylglycine-d4)
  • Glycochenodeoxycholic acid 3-sulfate disodium
  • Glycochenodeoxycholic acid 3-glucuronide
  • Glycochenodeoxycholic acid
Official Supplier of:
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Top Publications Citing lnvivochem Products
Product Description

Glycochenodeoxycholic acid sodium, the sodium salt of Glycochenodeoxycholic acid which is a glycine conjugate of lithocholic acid, is a bile salt produced in the liver from chenodeoxycholate and glycine.

Biological Activity I Assay Protocols (From Reference)
Targets
Endogenous Metabolite; Microbial Metabolite
ln Vitro
Cholestasis represents pathophysiologic syndromes defined as impaired bile flow from the liver. As an outcome, bile acids accumulate and promote hepatocyte injury, followed by liver cirrhosis and liver failure. Glycochenodeoxycholic acid (GCDCA) is relatively toxic and highly concentrated in bile and serum after cholestasis. However, the mechanism underlying GCDCA-induced hepatotoxicity remains unclear. In this study, we found that GCDCA inhibits autophagosome formation and impairs lysosomal function by inhibiting lysosomal proteolysis and increasing lysosomal pH, contributing to defects in autophagic clearance and subsequently leading to the death of L02 human hepatocyte cells. Notably, through tandem mass tag (TMT)-based quantitative proteomic analysis and database searches, 313 differentially expressed proteins were identified, of which 71 were increased and 242 were decreased in the GCDCA group compared with those in the control group. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that GCDCA suppressed the signaling pathway of transcription factor E3 (TFE3), which was the most closely associated with autophagic flux impairment. In contrast, GCDCA-inhibited lysosomal function and autophagic flux were efficiently attenuated by TFE3 overexpression. Specifically, the decreased expression of TFE3 was closely related to the disruption of reactive oxygen species (ROS) homeostasis, which could be prevented by inhibiting intracellular ROS with N-acetyl cysteine (NAC). In summary, our study is the first to demonstrate that manipulation of ROS/TFE3 signaling may be a therapeutic approach for antagonizing GCDCA-induced hepatotoxicity. [1]
Methods: TGF-β mRNA expression was documented in bile duct epithelial cells exposed to varying concentrations of the toxic bile acid; glycochenodeoxycholic acid (GCDCA) ± PC. Results: In these experiments, as well as in co-culture experiments where bile duct epithelial cells were cultured with peripheral blood mononuclear cells and myofibroblasts, TGF-β mRNA expression remained unaltered in the presence or absence of PC. Moreover, collagen type Iα1 mRNA expression by myofibroblasts also remained unaltered.[2]
The effect of Glycochenodeoxycholic acid/GCDC-induced apoptosis on PKC activity and PKC's role in GCDCA-induced hepatocyte apoptosis is unclear. The specific aims of this study were to determine if GCDC-induced apoptosis changed intracellular PKC activity and if modulation of PKC activity affected GCDC-induced hepatocyte apoptosis. Apoptosis was induced in isolated hepatocytes using GCDC. PKC activity was measured and specific PKC and calpain inhibitors were used to study the effects of PKC and calpain modulation on GCDC-induced apoptosis. After 4 h exposure, 50 microM GCDC induced apoptosis in 42% of hepatocytes. Intracellular PKC activity decreased to 44% of controls 2 h after exposure of hepatocytes to GCDC (p < 0.001). Pre-incubation of hepatocytes with the calpain protease inhibitor restored PKC activity in GCDC exposed hepatocytes to 91 +/- 5% of control cells. Pre-incubation of hepatocytes with a calpain inhibitor decreased GCDC-induced apoptosis as did pre-incubation with the PKC activating phorbol ester, PMA. The combination of calpain inhibition and PMA further reduced GCDC-induced apoptosis but caused low level hepatic apoptosis. Inhibition of PKC with chelerythrine also substantially reduced GCDC-induced hepatocyte apoptosis. GCDC-induced apoptosis is associated with decreases in total cellular PKC activity, which appear to be dependent on intracellular calpain-like protease activity. The combination of protease inhibition and phorbol ester pretreatment preserved total cellular PKC activity and decreased GCDC-induced apoptosis but induced low level apoptosis in the absence of GCDC exposure. PKC inhibition also decreased GCDC-induced hepatocyte apoptosis highlighting the complex interactions of PKC and proteases during GCDC-induced apoptosis. [3]
Glycochenodeoxycholic acid (GCDC), a component of bile acid (BA), has been reported to induce necrosis in primary human hepatocytes. In the present work, we investigated the function of GCDC in HCC chemoresistance. We found that GCDC promoted chemoresistance in HCC cells by down-regulating and up-regulating the expression of apoptotic and anti-apoptotic genes, respectively. Furthermore, GCDC induced the EMT phenotype and stemness in HCC cells and activated the STAT3 signaling pathway. These findings reveal that GCDC promotes chemoresistance in HCC by inducing stemness via the STAT3 pathway and could be a potential target in HCC chemotherapy [4].
ln Vivo
We then assessed whether direct injection of these selected bile acids into the ventricle of the brain could also suppress the HPA axis in vivo. The suppressive effects of bile acids on the HPA axis were restricted to the conjugated bile acids TCA and Glycochenodeoxycholic acid (GCDA), whereas the unconjugated bile acids CA, DCA, and CDCA had no significant effect on circulating corticosterone levels 6 hours after injection (Figure 3A). In parallel, hypothalamic CRH mRNA expression and circulating CRH protein levels were decreased by TCA and Glycochenodeoxycholic acid (GCDA) injection (Figure 3, B and C), whereas the bile acids CA, DCA, and CDCA had no significant effect (data not shown).[5]
To assess whether the in vivo effect of bile acids was also dependent upon ASBT, we injected an ASBT Vivo-Morpholino or mismatched control sequence into the third ventricle. This significantly suppressed the translation of ASBT protein in the hypothalamic neurons as demonstrated by immunofluorescence (Figure 5A). Direct injection of Glycochenodeoxycholic acid (GCDA) into the third ventricle significantly suppressed the hypothalamic CRH mRNA expression, (Figure 5B) protein content (Figure 5C) and circulating corticosterone levels (Figure 5D), an effect that was attenuated or reversed by ASBT Vivo-Morpholino injection.[5]
We then assessed whether the ability of bile acids to activate GR is also relevant in vivo. Rats were injected with GR Vivo-Morpholino (or mismatched control sequence) into the lateral ventricle, and the resulting expression of GR in the hypothalamus was assessed by immunofluorescence. As expected, GR immunoreactivity was significantly suppressed after central GR Vivo-Morpholino injection compared with the mismatched control as assessed by immunofluorescence (Figure 7A) and immunoblotting (Figure 7B). Furthermore, subsequent injection of Glycochenodeoxycholic acid (GCDA) into the third ventricle significantly suppressed the CRH mRNA expression (Figure 7C), protein content (Figure 7D), and circulating corticosterone levels (Figure 7E), an effect that was attenuated after GR Vivo-Morpholino injection.[5]
Enzyme Assay
GR luciferase assay [5]
In addition, the GR transcriptional activity was assessed in hypothalamic neurons using a luciferase reporter construct coupled to a promoter region containing the glucocorticoid response element (GRE) consensus sequence following procedures described previously. Neurons were plated onto 96-well plates at a density of 10 000 cells/well and allowed to adhere overnight. Cells were then transfected with the GRE-luciferase reporter construct (0.1-μg DNA/well) with 0.28 μL of TransIT-LT1 transfection reagent overnight at 37°C. After this time, the cells were stimulated with Glycochenodeoxycholic acid (GCDA) or TCA (10μM) and were assayed for luciferase activity using the luciferase assay kit 24 hours after stimulation. Treatments were done at least in quadruplicate, and results are expressed as the degree of change of luciferase activity per microgram of protein.
Cell Assay
Cell culture [1]
L02 human normal liver cells were cultured in 1640 medium (HyClone) supplemented with 10% heat-inactivated FCS and 1% (v/v) penicillin/streptomycin (Sigma, St Louis, MO, USA) in a 5% CO2 humidified atmosphere at 37 °C. At 80% confluence, the cells were treated with GCDCA at different concentrations (50, 75, or 100 μM) for 6 h or with 100 μM GCDCA for various periods (0, 1, 3, or 6 h), as described in our previous study (Chen et al., 2013, 2015; Xu et al., 2012). GCDCA was dissolved in sterile phosphate-buffered saline (PBS) to produce a 100 mM stock solution and then used to produce a serial dilution with cell culture medium before application.
Cell sample preparation and bile acid detection [1]
L02 cells(5 × 106 cells) were treated with 100 μMGCDCA for 6 h, and the cell samples were homogenized on ice in 500 μl of a mixture of chloroform, methanol and water (1:2.5:1, v/v/v). The samples were then centrifuged at 13,000 rpm for 10 min at 4 °C, and a 150-μl aliquot of the supernatant was transferred to an LC sampling vial containing an IS (10 μl L-4-chloro-phenylalanine in water, 5 μg/mL). The deposit was rehomogenized with 500 μl of methanol, and a 150-μl aliquot of supernatant was added to the same vial for drying prior to reconstitution with acetonitrile/H2O (6:4, v/v) to a final volume of 500 μl. After reconstituted with mobile phase, the extract as well as the bile acid reference standards were analyzed with a Waters ACQUITY ultra performance liquid chromatography coupled with a Waters XEVO TQ-S mass spectrometer with an ESI source. The entire UPLC–MS/MS system was controlled by MassLynx 4.1 software. All chromatographic separations were performed with an ACQUITY BEH C18 column (1.7 μm, 100 mm × 2.1 mm internal dimensions) and the injection volume was 5 μL. UPLC-MS raw data obtained with negative mode were analyzed using TargetLynx applications manager version 4.1 to obtain calibration equations and the quantitative concentration of each bile acid in the samples. Lysate samples were measured in ng/well and scaled per mg protein as measured using the Pierce BCA™ Protein Assay Kit.
Cell death assay [1]
L02 cells were plated in 6-well plates (5 × 105 cells per well). After being treated with GCDCA, the cells were detached with 300 μl of a trypsin EDTA solution. The suspension of detached cells was centrifuged at 300g for 5 min. Then, the pellet was combined with 800 μl of trypan blue solution and dispersed. After staining for 3 min, the cells were counted using an automated cell counter (Bio-Rad, TC10). The dead cells were stained blue. The cell mortality (%) is expressed as the percentage of dead cells/total cells (Chang et al., 2011).
Cell Culture [2]
KMBC cells were cultured in DMEM supplemented with 110 mg/L sodium pyruvate, 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 μg/ml streptomycin. LX-2 cells were cultured in DMEM/F12 supplemented with 0.1 mmol/L non-essential amino acid, 2 mmol/L glutamine, 110 mg/L sodium pyruvate, 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 μg/ml streptomycin. All the cells were incubated at 37 °C in a humidified atmosphere of 5% CO2 and 95% air. In experiments, where cells were exposed to GCDCA, the duration of exposure was 24 h.
Co-culture of KMBC and LX-2 Cells [2]
KMBC and LX-2 cells were cultured in co-culture plates with inserts containing a 3.0-um porous membrane at 37 °C in a humidified atmosphere of 5% CO2 and 95% air. For each plate, 8 × 106 KMBC cells were cultured in the upper chamber and 8 × 106 LX-2 cells in the lower chamber. Culture medium was removed one day before treatment and cells were incubated in the medium described above. The indicated concentrations of GCDCA were added into the upper chamber bath solution for 24 h prior to cell harvesting.
Co-culture of KMBC Peripheral Blood Mononuclear Cells (PBMC) and LX-2 Cells [2]
KMBC, PBMC (derived from healthy donors) and LX-2 cells were cultured in co-culture plates with inserts containing a 3.0-um porous membrane at 37 °C in a humidified atmosphere of 5% CO2 and 95% air. For each well, 5 × 106 KMBC and 3 × 106 PBMC cells were cultured in the upper chamber and 8 × 106 LX-2 cells in the lower chamber. Culture medium was removed one day before GCDCA exposure and the cells incubated in the medium described above.
Cell proliferation and cytotoxicity assay [4]
Chemotherapy-induced cell death was determined by cell counting kit-8 assay. Huh7 and LM3 cells were seeded in a 96-well plate at a density of 8 × 103 cells/well and incubated with GCDCA and treated with chemotherapeutic drugs (5-FU and cisplatin) for 24 h and 48 h, respectively. Next, the cells were washed with phosphate-buffered saline (PBS), and cell counting kit-8 (CCK-8) solution (1/10 the volume of media) was added for 1 h. The cell viability was detected at 450 nm using a microplate reader.
Apoptosis assay [4]
A total of 1 × 105 cells were seeded in a 6-well plate and treated with GCDCA and chemotherapeutic drugs for 24 h and 48 h, respectively. Next, the cells were washed with PBS and resuspended in the PBS and stained with Annexin V and propidium iodide (PI) according to manufacturer’s instructions. Flow cytometry was used to analyze the proportion of apoptotic cells.
Animal Protocol
Male Sprague Dawley rats (150–175 g) were maintained in a temperature-controlled environment (20°C–22°C) with a 12-hour light, 12-hour dark cycle. Unless otherwise indicated, animals had free access to drinking water and standard rat chow. Rats were fed a diet containing 2% cholestyramine or the control diet AIN-93G for 3 days before either BDL or sham surgeries. Tissue and serum were collected 3 days after surgery between the hours of 8 and 9 am to minimize the circadian variations in glucocorticoid levels. In a separate experiment, rats were injected with 20 pmol of the bile acids cholic acid (CA), deoxycholic acid (DCA), chenodeoxycholic acid (CDCA), Glycochenodeoxycholic acid (GCDA), or TCA in the third ventricle (0 mm medial/lateral, −1.8 mm anterior/posterior, +4.5 mm dorsal/ventral) and serum and tissue were collected 6 hours later. In parallel, rats were infused with 1 mg/kg · d of Vivo-Morpholino sequences into the lateral ventricle at the coordinates (−1.3 mm medial/lateral, −0.2 mm anterior/posterior, +3.5 mm dorsal/ventral) using the brain infusion kits coupled to subcutaneous implanted minipumps for 3 days before the single Glycochenodeoxycholic acid (GCDA) or TCA injection following the method described above. The degree by which the target gene expression was suppressed by Vivo-Morpholino infusion was evaluated by immunofluorescence and immunoblotting as previously described [5].
References

[1]. Glycochenodeoxycholic acid sodium salt impairs transcription factor E3 -dependent autophagy-lysosome machinery by disrupting reactive oxygen species homeostasis in L02 cells. Toxicol Lett. 2020 Oct 1;331:11-21.

[2]. Glycochenodeoxycholic acid sodium salt Does Not Increase Transforming Growth Factor-Beta Expression in Bile Duct Epithelial Cells or Collagen Synthesis in Myofibroblasts. J Clin Exp Hepatol. 2017 Dec;7(4):316-320.

[3]. Glycochenodeoxycholic acid (GCDC) induced hepatocyte apoptosis is associated with early modulation of intracellular PKC activity. Mol Cell Biochem. 2000 Apr;207(1-2):19-27.

[4]. Glycochenodeoxycholic acid sodium salt induces stemness and chemoresistance via the STAT3 signaling pathway in hepatocellular carcinoma cells. Aging (Albany NY). 2020 Aug 3;12(15):15546-15555.

[5]. Suppression of the HPA Axis During Cholestasis Can Be Attributed to Hypothalamic Bile Acid Signaling. Mol Endocrinol. 2015 Dec;29(12):1720-30.

Additional Infomation
Glycochenodeoxycholic acid is a bile acid glycine conjugate having 3alpha,7alpha-dihydroxy-5beta-cholan-24-oyl as the bile acid component. It has a role as a human metabolite. It is functionally related to a chenodeoxycholic acid. It is a conjugate acid of a glycochenodeoxycholate.
Glycochenodeoxycholic acid has been reported in Homo sapiens with data available.
A bile salt formed in the liver from chenodeoxycholate and glycine, usually as the sodium salt. It acts as a detergent to solubilize fats for absorption and is itself absorbed. It is a cholagogue and choleretic.
Recently, experimental evidence has indicated that deregulation of hepatic autophagic flux plays an important role in the pathogenesis of extrahepatic cholestasis. Impaired autophagy promoted bile acid-induced hepatic injury and the accumulation of ubiquitinated proteins, and activated of autophagy protected against cholestasis-induced hepatic injury (Gao et al., 2014; Kim et al., 2018). Moreover, Khambu B et al. reported that hepatic autophagy deficiency compromised farnesoid X receptor functionality and caused cholestatic injury (Khambu et al., 2019). Consistent with these findings, the results from our study confirmed that GCDCA inhibits autophagosome formation and impairs lysosomal function by inhibiting lysosomal proteolysis and increasing lysosomal pH, contributing to defects in autophagic clearance in vitro.
The bHLH-leucine zipper protein TFE3, which belongs to the MiTF/TFE family, is a master regulator of autophagy and lysosomal biogenesis and stimulates the overall degradation of cells (Fan et al., 2018). TFE3 is emerging as a global regulator of cell survival and energy metabolism, both through the promotion of lysosomal genes and through newly characterized targets, such as oxidative metabolism and the oxidative stress response (Pi et al., 2019a; Wang et al., 2019). More recently, other MiTF/TFE proteins, namely, melanocyte inducing transcription factor (MITF), transcription factor EB (TFEB) and transcription factor EC (TFEC), major regulators of autophagy and lysosomal biogenesis, have emerged as leading factors in human disease pathology (Martina et al., 2014). In our research, GCDCA treatment inhibited TFE3 expression and suppressed TFE3 reporter activity, which decreased the expression of autophagy-related genes. MITF, TFEB, and TFE3 show a ubiquitous pattern of expression and have been detected in multiple cell types, whereas TFEC expression is restricted to cells of myeloid origin (Slade and Pulinilkunnil, 2017). On the basis of these results, we investigated whether the other MiT/TFE proteins are involved in the action of GCDCA in L02 cells. Consistent with the proteomic analysis, TFE3 mRNA expression decreased significantly after exposure to different concentrations of GCDCA for 6 h, and no significant changes were detected in the levels of MITF or TFEB (Fig. S6). These results confirm the important role of TFE3 in GCDCA-mediated autophagy.
However, the mechanism that underlies the GCDCA-mediated inhibition of TFE3 expression and activity remains elusive. Recent studies have linked the accumulation of ROS to TFE3 activation in the invasion and migration of breast cancer or melanoma cells (Deng et al., 2018; Tan et al., 2018). ROS might play important roles in TFE3 inhibition in GCDCA-treated L02 cells. We found that GCDCA induced ROS generation in a dose-dependent manner, an effect abolished in cells pretreated with NAC. Furthermore, L02 cells incubated with NAC for 2 h prior to treatment with GCDCA showed inhibited ROS generation, which abrogated the effect of GCDCA on the TFE3 pathway. This result contradicts that of previous studies (Deng et al., 2018; Tan et al., 2018). We propose two possible reasons for this phenomenon. First, our results were obtained from a normal cell line, not a cancer cell line. Second, a very narrow spectrum of conditions was tested in the our study. The exact relationship between ROS and TFE3 may depend on the cell model, and the elucidation of the mechanistic details requires further research.
In summary, our data suggest that ROS/TFE3 signaling may serve as a therapeutic target for the development of novel treatments to prevent liver damage in patients with extrahepatic cholestasis (Fig. 7). Notwithstanding the above findings, a number of limitations of the study warrant emphasis. First, only one cell line was used to evaluate the mechanism of GDCDA-induced hepatotoxicity, and other hepatic cell lines and/or primary hepatocytes will be studied in our future work. Second, only GCDCA and no other toxic bile acids were studied. Most importantly, our results are from cultured cells, and we should be careful extrapolating results from in vitro culture experiments to human patient populations. The problems of the current system are expected to be overcome by further improvements, including through the use of animal studies and rigorous clinical trials in our future work.[1]
Notwithstanding the above findings, there are a number of limitations to the study that warrant emphasis. First, the cells employed included bile duct epithelial and myofibroblast cell lines rather than primary cells derived from human livers. Whether these cell lines are less sensitive to the toxic effects of GCDCA and pro-fibrogenic cytokine stimulation than primary cells remains to be determined. Second, only GCDCA and not other toxic bile acids were studied. Third, the concentration range of GCDCA was derived from previous reports documenting concentrations of GCDCA in human blood.14 Whether higher concentrations are present in human bile and in particular, the bile of PSC patients, is unclear. Fourth, the co-culture experiments physically separated bile duct epithelial and peripheral blood mononuclear cells from myofibroblasts thereby preventing cell–cell contact and possible intercellular communication. Fifth, perhaps longer periods of cell exposure to GCDCA were required to induce bile duct epithelial cell injury. However, the rapid biochemical and histologic changes associated with acute bile duct ligation models argue against that possibility.15 Sixth, it should be noted that PBMCs consist of peripheral blood monocytes and not tissue macrophages. Perhaps the additional features of the latter cell population are essential for the expression and release of pro-fibrogenic cytokines in this setting.16 Finally, we did not explore the possibility that restoring bile PC concentrations to normal levels may favorably alter the course of PSC by means other than protecting biliary tract epithelial cells from toxic bile acid-induced injury.
In conclusion, the results of this study do not support the hypothesis that PC deficiency permits toxic bile acid-induced injury of biliary tract epithelial cells and subsequent activation of adjacent myofibroblasts, resulting in the enhanced fibrosis seen in PSC. Thus, at this time, restoration of low PC levels to normal values in PSC bile does not appear to be a worthwhile therapeutic approach to the treatment of PSC. [2]
Studies have demonstrated that the JAK/STAT3 signaling pathway contributes to cell survival and chemotherapeutic resistance in cancers. Furthermore, it enhances the development of CSC-like characteristics. For example, the CSC marker, Nanog, is induced by the STAT3pathway in liver tumor-initiating cells. Similarly, members of the SOCS and PTPN families have been demonstrated to negatively affect the JAK/STAT signaling pathway. We found that GCDC activated the STAT3 signaling pathway by repressing the expression of several negative regulators of STAT3 signaling, including SOCS2, SOCS5, PTPN1, and PTPN11 in HCC cells. Glycochenodeoxycholic acid (GCDA)-induced resistance to drugs was inhibited when the expression of STAT3 was suppressed by siRNA in HCC cells. These results demonstrated that the STAT3 signaling pathway is involved in GCDC-induced chemoresistance of HCC cells. To summarize, our results showed that the treatment with GCDC enhanced the chemoresistance of HCC cells by inducing CSC-like characteristics and EMT phenotype, and activating the STAT3 signaling pathway via suppression of the expression of SOCS2, SOCS5, PTPN1, and PTPN1. Therefore, GCDC could serve as a potential target for the prognosis and therapy of HCC. [3]
Suppression of the hypothalamic-pituitary-adrenal (HPA) axis has been shown to occur during cholestatic liver injury. Furthermore, we have demonstrated that in a model of cholestasis, serum bile acids gain entry into the brain via a leaky blood brain barrier and that hypothalamic bile acid content is increased. Therefore, the aim of the current study was to determine the effects of bile acid signaling on the HPA axis. The data presented show that HPA axis suppression during cholestatic liver injury, specifically circulating corticosterone levels and hypothalamic corticotropin releasing hormone (CRH) expression, can be attenuated by administration of the bile acid sequestrant cholestyramine. Secondly, treatment of hypothalamic neurons with various bile acids suppressed CRH expression and secretion in vitro. However, in vivo HPA axis suppression was only evident after the central injection of the bile acids taurocholic acid or Glycochenodeoxycholic acid (GCDA) but not the other bile acids studied. Furthermore, we demonstrate that taurocholic acid and Glycochenodeoxycholic acid (GCDA) are exerting their effects on hypothalamic CRH expression after their uptake through the apical sodium-dependent bile acid transporter and subsequent activation of the glucocorticoid receptor. Taken together with previous studies, our data support the hypothesis that during cholestatic liver injury, bile acids gain entry into the brain, are transported into neurons through the apical sodium-dependent bile acid transporter and can activate the glucocorticoid receptor to suppress the HPA axis. These data also lend themselves to the broader hypothesis that bile acids may act as central modulators of hypothalamic peptides that may be altered during liver disease.[5]
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C26H42NNAO5
Molecular Weight
471.6052
Exact Mass
471.296
CAS #
16564-43-5
Related CAS #
Glycochenodeoxycholic acid;640-79-9;Glycochenodeoxycholic acid-d7 sodium;Glycochenodeoxycholic acid-d4;1201918-16-2
PubChem CID
12544
Appearance
White to off-white solid powder
Boiling Point
655.6ºC at 760mmHg
Flash Point
350.3ºC
Vapour Pressure
5.74E-20mmHg at 25°C
LogP
2.65
Hydrogen Bond Donor Count
4
Hydrogen Bond Acceptor Count
5
Rotatable Bond Count
6
Heavy Atom Count
32
Complexity
727
Defined Atom Stereocenter Count
10
SMILES
C[C@H](CCC(=O)NCC(=O)O)[C@H]1CC[C@@H]2[C@@]1(CC[C@H]3[C@H]2[C@@H](C[C@H]4[C@@]3(CC[C@H](C4)O)C)O)C
InChi Key
GHCZAUBVMUEKKP-GYPHWSFCSA-N
InChi Code
InChI=1S/C26H43NO5/c1-15(4-7-22(30)27-14-23(31)32)18-5-6-19-24-20(9-11-26(18,19)3)25(2)10-8-17(28)12-16(25)13-21(24)29/h15-21,24,28-29H,4-14H2,1-3H3,(H,27,30)(H,31,32)/t15-,16+,17-,18-,19+,20+,21-,24+,25+,26-/m1/s1
Chemical Name
2-[[(4R)-4-[(3R,5S,7R,8R,9S,10S,13R,14S,17R)-3,7-dihydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-17-yl]pentanoyl]amino]acetic acid
Synonyms
16564-43-5; Glycochenodeoxycholic acid sodium salt; Sodium glycochenodeoxycholate; Sodium glycylchenodeoxycholate; NSC 681056; CHENYLGLYCINE SODIUM; Glycochenodeoxycholic acid (sodium salt); OK5NH65A9B;
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: Please store this product in a sealed and protected environment, avoid exposure to moisture.
Shipping Condition
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
Solubility Data
Solubility (In Vitro)
DMSO : ~250 mg/mL (~530.10 mM)
H2O : ≥ 100 mg/mL (~212.04 mM)
Solubility (In Vivo)
Solubility in Formulation 1: ≥ 2.08 mg/mL (4.41 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 20.8 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: ≥ 2.08 mg/mL (4.41 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 20.8 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: ≥ 2.08 mg/mL (4.41 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (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 20.8 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly.


Solubility in Formulation 4: 50 mg/mL (106.02 mM) in PBS (add these co-solvents sequentially from left to right, and one by one), clear solution; with ultrasonication.

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 2.1204 mL 10.6020 mL 21.2040 mL
5 mM 0.4241 mL 2.1204 mL 4.2408 mL
10 mM 0.2120 mL 1.0602 mL 2.1204 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.

Calculator

Molarity Calculator allows you to calculate the mass, volume, and/or concentration required for a solution, as detailed below:

  • Calculate the Mass of a compound required to prepare a solution of known volume and concentration
  • Calculate the Volume of solution required to dissolve a compound of known mass to a desired concentration
  • Calculate the Concentration of a solution resulting from a known mass of compound in a specific volume
An example of molarity calculation using the molarity calculator is shown below:
What is the mass of compound required to make a 10 mM stock solution in 5 ml of DMSO given that the molecular weight of the compound is 350.26 g/mol?
  • Enter 350.26 in the Molecular Weight (MW) box
  • Enter 10 in the Concentration box and choose the correct unit (mM)
  • Enter 5 in the Volume box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 17.513 mg appears in the Mass box. In a similar way, you may calculate the volume and concentration.

Dilution Calculator allows you to calculate how to dilute a stock solution of known concentrations. For example, you may Enter C1, C2 & V2 to calculate V1, as detailed below:

What volume of a given 10 mM stock solution is required to make 25 ml of a 25 μM solution?
Using the equation C1V1 = C2V2, where C1=10 mM, C2=25 μM, V2=25 ml and V1 is the unknown:
  • Enter 10 into the Concentration (Start) box and choose the correct unit (mM)
  • Enter 25 into the Concentration (End) box and select the correct unit (mM)
  • Enter 25 into the Volume (End) box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 62.5 μL (0.1 ml) appears in the Volume (Start) box
g/mol

Molecular Weight Calculator allows you to calculate the molar mass and elemental composition of a compound, as detailed below:

Note: Chemical formula is case sensitive: C12H18N3O4  c12h18n3o4
Instructions to calculate molar mass (molecular weight) of a chemical compound:
  • To calculate molar mass of a chemical compound, please enter the chemical/molecular formula and click the “Calculate’ button.
Definitions of molecular mass, molecular weight, molar mass and molar weight:
  • Molecular mass (or molecular weight) is the mass of one molecule of a substance and is expressed in the unified atomic mass units (u). (1 u is equal to 1/12 the mass of one atom of carbon-12)
  • Molar mass (molar weight) is the mass of one mole of a substance and is expressed in g/mol.
/

Reconstitution Calculator allows you to calculate the volume of solvent required to reconstitute your vial.

  • Enter the mass of the reagent and the desired reconstitution concentration as well as the correct units
  • Click the “Calculate” button
  • The answer appears in the Volume (to add to vial) box
In vivo Formulation Calculator (Clear solution)
Step 1: Enter information below (Recommended: An additional animal to make allowance for loss during the experiment)
Step 2: Enter in vivo formulation (This is only a calculator, not the exact formulation for a specific product. Please contact us first if there is no in vivo formulation in the solubility section.)
+
+
+

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.

Contact Us