Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Edith K.Y. Tang is active.

Publication


Featured researches published by Edith K.Y. Tang.


The FASEB Journal | 2012

In vivo evidence for a novel pathway of vitamin D3 metabolism initiated by P450scc and modified by CYP27B1

Andrzej Slominski; Tae Kang Kim; Haleem Z. Shehabi; Igor Semak; Edith K.Y. Tang; Minh N. Nguyen; Heather A. E. Benson; Elena Korik; Zorica Janjetovic; Jianjun Chen; Charles R. Yates; Arnold E. Postlethwaite; Wei Li; Robert C. Tuckey

We define previously unrecognized in vivo pathways of vitamin D3 (D3) metabolism generating novel D3‐hydroxyderivatives different from 25‐hydroxyvitamin D3 [25(OH)D3] and 1,25(OH)2D3. Their novel products include 20‐hydroxyvitamin D3 [20(OH)D3], 22(OH)D3, 20,23(OH)2D3, 20,22(OH)2D3, 1,20(OH)2D3,1,20,23(OH)3D3, and 17,20,23(OH)3D3 and were produced by placenta, adrenal glands, and epidermal keratinocytes. We detected the predominant metabolite [20(OH)D3] in human serum with a relative concentration ~20 times lower than 25(OH)D3. Use of inhibitors and studies performed with isolated mitochondria and purified enzymes demonstrated involvement of the steroidogenic enzyme cytochrome P450scc (CYP11A1) as well as CYP27B1 (1α‐hydroxylase). In placenta and adrenal glands with high CYP11A1 expression, the predominant pathway was D3 → 20(OH)D3 → 20,23(OH)2D3 → 17,20,23(OH)3D3 with further 1α‐hydroxylation, and minor pathways were D3 → 25(OH)D3 → 1,25(OH)2D3 and D3 → 22(OH)D3 → 20,22(OH)2D3. In epidermal keratinocytes, we observed higher proportions of 22(OH)D3 and 20,22(OH)2D3. We also detected endogenous production of 20(OH)D3, 22(OH) D3, 20,23(OH)2D3, 20,22(OH)2D3, and 17,20,23(OH)3D3 by immortalized human keratinocytes. Thus, we provide in vivo evidence for novel pathways of D3 metabolism initiated by CYP11A1, with the product profile showing organ/cell type specificity and being modified by CYP27B1 activity. These findings define the pathway intermediates as natural products/endogenous bioregulators and break the current dogma that vitamin D is solely activated through the sequence D3 → 25(OH)D3 → 1,25(OH)2D3.—Slominski, A. T., Km, T.‐K., Shehabi, H. Z., Semak, I., Tang, E. K. Y., Nguyen, M. N., Benson, H. A. E., Korik, E., Janjetovic, Z., Chen, J., Yates, C. R., Postlethwaite, A., Li, W., Tuckey, R. C. In vivo evidence for a novel pathway of vitamin D3 metabolism initiated by P450scc and modified by CYP27B1. FASEB J. 26, 3901–3915 (2012). www.fasebj.org


American Journal of Physiology-cell Physiology | 2011

20-Hydroxyvitamin D2 is a noncalcemic analog of vitamin D with potent antiproliferative and prodifferentiation activities in normal and malignant cells

Andrzej Slominski; Tae Kang Kim; Zorica Janjetovic; Robert C. Tuckey; Radoslaw Bieniek; Junming Yue; Wei Li; Jianjun Chen; Minh N. Nguyen; Edith K.Y. Tang; Duane D. Miller; Tai C. Chen; Michael F. Holick

20-hydroxyvitamin D(2) [20(OH)D(2)] inhibits DNA synthesis in epidermal keratinocytes, melanocytes, and melanoma cells in a dose- and time-dependent manner. This inhibition is dependent on cell type, with keratinocytes and melanoma cells being more sensitive than normal melanocytes. The antiproliferative activity of 20(OH)D(2) is similar to that of 1,25(OH)(2)D(3) and of newly synthesized 1,20(OH)(2)D(2) but significantly higher than that of 25(OH)D(3). 20(OH)D(2) also displays tumorostatic effects. In keratinocytes 20(OH)D(2) inhibits expression of cyclins and stimulates involucrin expression. It also stimulates CYP24 expression, however, to a significantly lower degree than that by 1,25(OH)(2)D(3) or 25(OH)D(3). 20(OH)D(2) is a poor substrate for CYP27B1 with overall catalytic efficiency being 24- and 41-fold lower than for 25(OH)D(3) with the mouse and human enzymes, respectively. No conversion of 20(OH)D(2) to 1,20(OH)(2)D(2) was detected in intact HaCaT keratinocytes. 20(OH)D(2) also demonstrates anti-leukemic activity but with lower potency than 1,25(OH)(2)D(3). The phenotypic effects of 20(OH)D(2) are mediated through interaction with the vitamin D receptor (VDR) as documented by attenuation of cell proliferation after silencing of VDR, by enhancement of the inhibitory effect through stable overexpression of VDR and by the demonstration that 20(OH)D(2) induces time-dependent translocation of VDR from the cytoplasm to the nucleus at a comparable rate to that for 1,25(OH)(2)D(3). In vivo tests show that while 1,25(OH)(2)D(3) at doses as low as 0.8 μg/kg induces calcium deposits in the kidney and heart, 20(OH)D(2) is devoid of such activity even at doses as high as 4 μg/kg. Silencing of CY27B1 in human keratinocytes showed that 20(OH)D(2) does not require its transformation to 1,20(OH)(2)D(2) for its biological activity. Thus 20(OH)D(2) shows cell-type dependent antiproliferative and prodifferentiation activities through activation of VDR, while having no detectable toxic calcemic activity, and is a poor substrate for CYP27B1.


Scientific Reports | 2015

Detection of novel CYP11A1-derived secosteroids in the human epidermis and serum and pig adrenal gland.

Andrzej Slominski; Tae Kang Kim; Wei Li; Arnold E. Postlethwaite; Elaine W. Tieu; Edith K.Y. Tang; Robert C. Tuckey

To investigate whether novel pathways of vitamin D3 (D3) and 7-dehydrocholesterol (7DHC) metabolism initiated by CYP11A1 and previously characterized in vitro, occur in vivo, we analyzed samples of human serum and epidermis, and pig adrenals for the presence of intermediates and products of these pathways. We extracted human epidermis from 13 individuals and sera from 13 individuals and analyzed them by LC/qTOF-MS alongside the corresponding standards. Pig adrenal glands were also analyzed for these steroids and secosteroids. Epidermal, serum and adrenal samples showed the presence of D3 hydroxy-derivatives corresponding to 20(OH)D3, 22(OH)D3, 25(OH)D3, 1,25(OH)2D3, 20,22(OH)2D3, 20,23(OH)2D3, 20,24(OH)2D3, 20,25(OH)2D3, 20,26(OH)2D3, 1,20,23(OH)3D3 and 17,20,23(OH)3D3, plus 1,20(OH)2D3 which was detectable only in the epidermis. Serum concentrations of 20(OH)D3 and 22(OH)D3 were only 30- and 15-fold lower than 25(OH)D3, respectively, and at levels above those required for biological activity as measured in vitro. We also detected 1,20,24(OH)3D3, 1,20,25(OH)3D3 and 1,20,26(OH)3D3 in the adrenals. Products of CYP11A1 action on 7DHC, namely 22(OH)7DHC, 20,22(OH)27DHC and 7-dehydropregnenolone were also detected in serum, epidermis and the adrenal. Thus, we have detected novel CYP11A1-derived secosteroids in the skin, serum and adrenal gland and based on their concentrations and biological activity suggest that they act as hormones in vivo.


Molecular and Cellular Endocrinology | 2012

Correlation between secosteroid induced vitamin D receptor activity in melanoma cells and computer-modeled receptor binding strength

Tae-Kang Kim; Jin Wang; Zorica Janjetovic; Jianjun Chen; Robert C. Tuckey; Minh N. Nguyen; Edith K.Y. Tang; Duane D. Miller; Wei Li; Andrzej Slominski

To define the interaction of novel secosteroids produced by the action of cytochrome P450scc with vitamin D receptor (VDR), we used a human melanoma line overexpressing VDR fused with enhanced green fluorescent protein (EGFP) and tested the ligand induced translocation of VDR from the cytoplasm to the nucleus. Hydroxyderivatives of vitamin D(3) with a full length (D(3)) side chain and hydroxy-secosteroids with a shortened side chain (pD) stimulated VDR translocation and inhibited proliferation, however, with different potencies. In general the D(3) were more potent than pD analogues. Molecular modeling of the binding of the secosteroids to the VDR genomic binding pocket (G-pocket) correlated well with the experimental data for VDR translocation. In contrast, docking scores for the non-genomic binding site of the VDR were poor. In conclusion, both the length of the side chain and the number and position of hydroxyl groups affect the activation of VDR by novel secosteroids.


Molecular and Cellular Endocrinology | 2014

In vivo production of novel vitamin D2 hydroxy-derivatives by human placentas, epidermal keratinocytes, Caco-2 colon cells and the adrenal gland

Andrzej Slominski; Tae Kang Kim; Haleem Z. Shehabi; Edith K.Y. Tang; Heather A. E. Benson; Igor Semak; Zongtao Lin; Charles R. Yates; Jin Wang; Wei Li; Robert C. Tuckey

We investigated the metabolism of vitamin D2 to hydroxyvitamin D2 metabolites ((OH)D2) by human placentas ex-utero, adrenal glands ex-vivo and cultured human epidermal keratinocytes and colonic Caco-2 cells, and identified 20(OH)D2, 17,20(OH)₂D2, 1,20(OH)₂D2, 25(OH)D2 and 1,25(OH)₂D2 as products. Inhibition of product formation by 22R-hydroxycholesterol indicated involvement of CYP11A1 in 20- and 17-hydroxylation of vitamin D2, while use of ketoconazole indicated involvement of CYP27B1 in 1α-hydroxylation of products. Studies with purified human CYP11A1 confirmed the ability of this enzyme to convert vitamin D2 to 20(OH)D2 and 17,20(OH)₂D2. In placentas and Caco-2 cells, production of 20(OH)D2 was higher than 25(OH)D2 while in human keratinocytes the production of 20(OH)D2 and 25(OH)D2 were comparable. HaCaT keratinocytes showed high accumulation of 1,20(OH)₂D2 relative to 20(OH)D2 indicating substantial CYP27B1 activity. This is the first in vivo evidence for a novel pathway of vitamin D2 metabolism initiated by CYP11A1 and modified by CYP27B1, with the product profile showing tissue- and cell-type specificity.


The Journal of Steroid Biochemistry and Molecular Biology | 2010

Metabolism of substrates incorporated into phospholipid vesicles by mouse 25-hydroxyvitamin D3 1α-hydroxylase (CYP27B1)

Edith K.Y. Tang; Kimberley J.Q. Voo; Minh N. Nguyen; Robert C. Tuckey

CYP27B1 catalyzes the 1alpha-hydroxylation of 25-hydroxyvitamin D3 to 1alpha,25-dihydroxyvitamin D3, the hormonally active form of vitamin D3. To further characterize mouse CYP27B1, it was expressed in Escherichia coli, purified and its activity measured on substrates incorporated into phospholipid vesicles, which served as a model of the inner mitochondrial membrane. 25-Hydroxyvitamin D3 and 25-hydroxyvitamin D2 in vesicles underwent 1alpha-hydroxylation with similar kinetics, the catalytic rate constants (k(cat)) were 41 and 48mol/min/mol P450, respectively, while K(m) values were 5.9 and 4.6mmol/mol phospholipid, respectively. CYP27B1 showed inhibition when substrate concentrations in the membrane were greater than 4 times K(m), more pronounced with 25-hydroxyvitamin D3 than 25-hydroxyvitamin D2. Higher catalytic efficiency was seen in vesicles prepared from dioleoyl phosphatidylcholine and cardiolipin than for dimyristoyl phosphatidylcholine vesicles. CYP27B1 also catalyzed 1alpha-hydroxylation of vesicle-associated 24R,25-dihydroxyvitamin D3 and 20-hydroxyvitamin D3, and 25-hydroxylation of 1alpha-hydroxyvitamin D3 and 1alpha-hydroxyvitamin D2, but with much lower efficiency than for 25(OH)D3. This study shows that CYP27B1 can hydroxylate 25-hydroxyvitamin D2 and 25-hydroxyvitamin D3 associated with phospholipid membranes with the highest activity yet reported for the enzyme. The expressed enzyme has low activity at higher concentrations of 25-hydroxyvitamin D in membranes, revealing that substrate inhibition may contribute to the regulation of the activity of this enzyme.


Biochemical Pharmacology | 2012

Rat CYP24A1 acts on 20-hydroxyvitamin D3 producing hydroxylated products with increased biological activity

Elaine W. Tieu; Edith K.Y. Tang; Jianjun Chen; Wei Li; Minh N. Nguyen; Zorica Janjetovic; Andrzej Slominski; Robert C. Tuckey

20-Hydroxyvitamin D(3) (20(OH)D(3)), the major product of CYP11A1 action on vitamin D(3), is biologically active and is produced in vivo. As well as potentially having important physiological actions, it is of interest as a therapeutic agent due to its lack of calcemic activity. In the current study we have examined the ability of CYP24A1, the enzyme that inactivates 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)), to metabolize 20(OH)D(3). Rat CYP24A1 was expressed in Escherichia coli, purified by Ni-affinity chromatography and assayed with substrates incorporated into phospholipid vesicles which served as a model of the inner mitochondrial membrane. In this system CYP24A1 metabolized 1,25(OH)(2)D(3) with a catalytic efficiency 1.4-fold higher than that seen for 25-hydroxyvitamin D(3) (25(OH)D(3)). CYP24A1 hydroxylated 20(OH)D(3) to several dihydroxy-derivatives with the major two identified by NMR as 20,24-dihydroxyvitamin D(3) (20,24(OH)(2)D(3)) and 20,25-dihydroxyvitamin D(3) (20,25(OH)(2)D(3)). The catalytic efficiency of CYP24A1 for 20(OH)D(3) metabolism was more than 10-fold lower than for either 25(OH)D(3) or 1,25(OH)(2)D(3) and no secondary metabolites were produced. The two major products, 20,24(OH)(2)D(3) and 20,25(OH)(2)D(3), caused significantly greater inhibition of colony formation by SKMEL-188 melanoma cells than either 1,25(OH)(2)D(3) or the parent 20(OH)D(3), showing that CYP24A1 plays an activating, rather than an inactivating role on 20(OH)D(3).


Drug Metabolism and Disposition | 2010

Purified Mouse CYP27B1 Can Hydroxylate 20,23-Dihydroxyvitamin D3, Producing 1α,20,23-Trihydroxyvitamin D3, Which Has Altered Biological Activity

Edith K.Y. Tang; Wei Li; Zorica Janjetovic; Minh N. Nguyen; Zhao Wang; Andrzej Slominski; Robert C. Tuckey

20,23-Dihydroxyvitamin D3 [20,23(OH)2D3] is a biologically active metabolite produced by the action of cytochrome P450scc (CYP11A1) on vitamin D3. It inhibits keratinocyte proliferation, stimulates differentiation, and inhibits nuclear factor-κB activity, working as a vitamin D receptor agonist. We have tested the ability of purified mouse 25-hydroxyvitamin D3 1α-hydroxylase (CYP27B1) to add a 1α-hydroxyl group to this vitamin D analog and determined whether this altered its biological activity. 20,23(OH)2D3 incorporated into phospholipid vesicles was converted to a single product by CYP27B1, confirmed to be 1α,20,23-trihydroxyvitamin D3 [1,20,23(OH)3D3] by mass spectrometry and NMR. The 20,23(OH)2D3 was a relatively poor substrate for CYP27B1 compared with the normal substrate, 25-hydroxyvitamin D3, displaying a 5-fold higher Km and 8-fold lower kcat value. Both 20,23(OH)2D3 and 1,20,23(OH)3D3 decreased neonatal human epidermal keratinocyte proliferation, showing significant effects at a lower concentration (0.1 nM) than that seen for 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3] at 24 h of treatment. Both compounds also decreased cell biomass relative to that of control cells, measured by staining with sulforhodamine B. They caused little stimulation of the expression of the vitamin D receptor at the mRNA level compared with the 30-fold induction observed with the same concentration (100 nM) of 1,25(OH)2D3 at 24 h. Addition of a 1α-hydroxyl group to 20,23(OH)2D3 greatly enhanced its ability to stimulate the expression of the CYP24 gene but not to the extent seen with 1,25(OH)2D3. This study shows that purified CYP27B1 can add a 1α-hydroxyl group to 20,23(OH)2D3 with the product showing altered biological activity, especially for the stimulation of CYP24 gene expression.


Journal of Medicinal Chemistry | 2012

Design, Synthesis, and Biological Action of 20R-Hydroxyvitamin D3

Yan Lu; Jianjun Chen; Zorica Janjetovic; Phillip Michaels; Edith K.Y. Tang; Jin Wang; Robert C. Tuckey; Andrzej Slominski; Wei Li; Duane D. Miller

The non-naturally occurring 20R epimer of 20-hydroxyvitamin D3 is synthesized based on chemical design and hypothesis. The 20R isomer is separated by semipreparative HPLC, and its structure is characterized. A comparison of 20R isomer to its 20S counterpart in biological evaluation demonstrates that they have different behaviors in antiproliferative and metabolic studies.


FEBS Journal | 2014

Kinetic analysis of human CYP24A1 metabolism of vitamin D via the C24-oxidation pathway.

Elaine W. Tieu; Edith K.Y. Tang; Robert C. Tuckey

CYP24A1 is the multicatalytic cytochrome P450 responsible for the catabolism of vitamin D via the C23‐ and C24‐oxidation pathways. We successfully expressed the labile human enzyme in Escherichia coli and partially purified it in an active state that permitted detailed characterization of its metabolism of 1,25‐dihydroxyvitamin D3 [1,25(OH)2D3] and the intermediates of the C24‐oxidation pathway in a phospholipid‐vesicle reconstituted system. The C24‐oxidation pathway intermediates, 1,24,25‐trihydroxyvitamin D3, 24‐oxo‐1,25‐dihydroxyvitamin D3, 24‐oxo‐1,23,25‐trihydroxyvitamin D3 and tetranor‐1,23‐dihydroxyvitamin D3, were enzymatically produced from 1,25(OH)2D3 using rat CYP24A1. Both 1,25(OH)2D3 and 1,23‐dihydroxy‐24,25,26,27‐tetranorvitamin D3 were found to partition strongly into the phospholipid bilayer when in aqueous medium. Changes to the phospholipid concentration did not affect the kinetic parameters for the metabolism of 1,25(OH)2D3 by CYP24A1, indicating that it is the concentration of substrates in the membrane phase (mol substrate·mol phospholipid−1) that determines their rate of metabolism. CYP24A1 exhibited Km values for the different C24‐intermediates ranging from 0.34 to 15 mmol·mol phospholipid−1, with 24‐oxo‐1,23,25‐trihydroxyvitamin D3 [24‐oxo‐1,23,25(OH)3D3] displaying the lowest and 1,24,25‐trihydroxyvitamin D3 [1,24,25(OH)3D3] displaying the highest. The kcat values varied by up to 3.8‐fold, with 1,24,25(OH)3D3 displaying the highest kcat (34 min−1) and 24‐oxo‐1,23,25(OH)3D3 the lowest. The data show that the cleavage of the side chain of 24‐oxo‐1,23,25(OH)3D3 occurs with the highest catalytic efficiency (kcat/Km) and produces 1‐hydroxy‐23‐oxo‐24,25,26,27‐tetranorvitamin D3 and not 1,23‐dihydroxy‐24,25,26,27‐tetranorvitamin D3, as the primary product. These kinetic analyses also show that intermediates of the C24‐oxidation pathway effectively compete with precursor substrates for binding to the active site of the enzyme, which manifests as an accumulation of intermediates, indicating that they dissociate after each catalytic step.

Collaboration


Dive into the Edith K.Y. Tang's collaboration.

Top Co-Authors

Avatar

Robert C. Tuckey

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Andrzej Slominski

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Wei Li

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Zorica Janjetovic

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Jianjun Chen

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Elaine W. Tieu

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Minh N. Nguyen

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Tae Kang Kim

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Duane D. Miller

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge