Network


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

Hotspot


Dive into the research topics where Kyle W. Selcer is active.

Publication


Featured researches published by Kyle W. Selcer.


Steroids | 1998

Development of potent non-estrogenic estrone sulfatase inhibitors

Pui-Kai Li; Guo-Hua Chu; JingPing Guo; Amy Peters; Kyle W. Selcer

Estrogen levels in breast tumors of post-menopausal women are as much as 10 times higher than estrogen levels in plasma, presumably due to in situ formation of estrogen. The major source of estrogen in breast cancer cells may be conversion of estrone sulfate to estrone by the enzyme estrone sulfatase. Thus, inhibitors of estrone sulfatase have potential for the treatment of estrogen-dependent breast cancers. Several steroidal agents have been developed that are potent estrone sulfatase inhibitors, most notably estrone-3-O-sulfamate. These compounds may have undesired actions, especially estrogenicity. Recently, non-steroidal estrone sulfatase inhibitors have been designed that avoid the problems associated with an active steroid nucleus; however, these have not achieved the potency of estrone-3-O sulfamate. We have designed and synthesized a series of compounds, 17 beta-(N-alkylcarbamoyl)-estra-1,3,5(10)-trien-3-O-sulfamates (6a-d) and 17 beta-(N-alkanoyl)-estra-1,3,5(10)-trien-3-O-sulfamates (11a-d) that combine the structural features of the steroidal estrone sulfatase inhibitors with a membrane insertion region that should increase the affinity for the sulfatase enzyme and decrease the estrogenicity of the steroid. We tested the compounds for estrone sulfatase inhibition by measuring estrone sulfatase activity in intact cultures of human breast cancer cells (MDA-MB-231). We tested for estrogenicity by measuring growth of estrogen-dependent MCF-7 human breast cancer cells. All of the test compounds (10 nM) substantially inhibited estrogen sulfatase activity of intact MDA-MB-231 cells. Dose-response analysis indicated an IC50 of approximately 0.5 nM for two of the compounds (6a and 11a). In the test for estrogenicity, estrone and estrone-3-O-sulfamate significantly stimulated MCF-7 cell growth. In contrast, neither the 17 beta-(N-alkylcarbamoyl)-estra-1,3,5,(10)-trien-3-O-sulfamates++ + nor the 17 beta-(N)-alkanoyl)-estra-1,3,5,(10)-trien-3-O-sulfamates stimulated growth of MCF-7 cells at a concentration of 1 microM, indicating that they are not estrogenic at levels 2000 times greater than their IC50 for estrone sulfatase. Our data indicate the utility of the new compounds for inhibition of breast cancer cell estrone sulfatase activity. Further, our data support the concept that estrone sulfatase inhibitors may be useful as therapeutic agents for estrogen-dependent breast cancers.


Copeia | 1986

Life history of a successful colonizer: the mediterranean gecko, Hemidactylus turcicus, in southern Texas

Kyle W. Selcer

A southern Texas population of the introduced lizard Hemidactylus turcicus, was examined for life history characteristics of a colonizer. Density estimates were high, ranging from 544-2210 lizards per ha. The population was early maturing (8.6 months) with low fecundity (1-3 clutches of two eggs/yr) and high survivorship for a small lizard (55% annual turnover, three or more year lifespan). Survivorship was similar between adults and juveniles, but small juveniles (<30 mm) had significantly lower survivorship than large juveniles (30-43 mm). The Mediterranean geckos success in southern Texas appears to result from low predation pressure, little interspecific competition and a life history which maximizes survival at all ages. This geckos ability to disperse may be enhanced by its calcareous shelled eggs serving as propagules of new colonies.


The Journal of Steroid Biochemistry and Molecular Biology | 1996

Inhibition of placental estrone sulfatase activity and MCF-7 breast cancer cell proliferation by estrone-3-amino derivatives

Kyle W. Selcer; Sharada Jagannathan; Michael E. Rhodes; Pui-Kai Li

Estrogen levels in breast tumors of post-menopausal women are as much as 10 times higher than in plasma, presumably due to in situ formation of estrogen. Several lines of evidence indicate that the major source of estrogen in breast cancer cells may be from conversion of estrone sulfate to estrone by the enzyme estrone sulfatase. Inhibitors of estrone sulfatase may thus be potential agents for the treatment of estrogen-dependent breast cancer. We designed and synthesized a series of estrone-3-amino derivatives as potential estrone sulfatase inhibitors. We tested the inhibitory potential of these compounds using human placental microsomes, which contain a substantial amount of estrone sulfatase activity. Several compounds in the series significantly inhibited estrone sulfatase activity of the human placental microsomes when present at 10 microM. The IC50 for the estrone-3-amino compounds ranged from 8.7 to 14.6 microM. We next tested the ability of the estrone-3-amino derivatives to inhibit growth of the estrogen-dependent MCF-7 breast cancer cell line. MCF-7 cells showed substantial proliferation in the presence of 100 nM estrone sulfate in estrogen-free media, indicating that the cells were capable of converting estrone sulfate into estrone. The proliferative effect of estrone sulfate (1 microM) was significantly blocked by the estrone-3-amino derivatives at 10 microM. The magnitude of MCF-7 cell inhibition resulting from treatment with the estrone-3 amino compounds was similar to or exceeded that of Danazol, but was less than the level resulting from treatment with estrone sulfamate. Using data from all of the compounds tested, inhibition of MCF-7 cell proliferation was positively correlated with inhibition of placental estrone sulfatase activity, suggesting that the reduction in cell growth was attributable to the blockade of sulfatase activity. In support of this, there was no relationship between inhibition of estrone sulfatase activity and inhibition of cell growth when the estrogen-independent cell line MDA-MB-231 was used. Our results indicate the possible utility of estrone-3-amino derivatives for inhibition of estrone sulfatase activity. Further, our data support the concept that estrone sulfatase inhibitors may be useful as therapeutic agents for estrogen-dependent breast cancers.


Steroids | 2002

Inhibition of steryl sulfatase activity in LNCaP human prostate cancer cells

Kyle W. Selcer; Heidi L Kabler; Jennifer Sarap; Zili Xiao; Pui-Kai Li

The enzyme steryl sulfatase may help support the growth of hormone-dependent tumors, including prostate cancers, by facilitating the conversion of circulating precursor steroids to active hormones. We sought to determine the presence of steryl sulfatase activity in the androgen-dependent human prostate cancer cell line LNCaP, and to determine if this activity was inhibited by known steryl sulfatase inhibitors. Intact LNCaP cultures had steryl sulfatase activity, as determined by conversion of [3H]estrone sulfate (E(1)S) to unconjugated steroids. The level of steryl sulfatase activity was relatively low (4.6 pmol/18 h/million cells) compared to MDA-MB-231 breast cancer cells (284.0 pmol/18 h/million cells). The observed activity in both cell lines was blocked by addition of 1 microM estrone sulfamate (EMATE), an active-site-directed, steroidal inhibitor of steryl sulfatase. Steryl sulfatase activity was also inhibited by Danazol, and by (p-O-sulfamoyl)-tetradecanoyl tyramine (C2-14), a non-steroidal inhibitor. Microsomes prepared from LNCaP cultures also showed steryl sulfatase activity, as determined by hydrolysis of [3H]E(1)S and [3H]dehydroepiandrosterone sulfate (DHEAS) to unconjugated forms. LNCaP and MDA-MB-231 microsomes both hydrolyzed E(1)S about two times faster than DHEAS. Hydrolysis of E(1)S in LNCaP and MDA-MB-231 microsomes was blocked by steryl sulfatase inhibitors with the following relative potencies: EMATE>C2-14>Danazol. These data demonstrate that LNCaP prostate cancer cells contain a steryl sulfatase with properties similar to that found in human breast cancer cells, and that the activity of this enzyme can be blocked by known steryl sulfatase inhibitors. Steryl sulfatase inhibitors may be useful as an adjuvant to androgen deprivation therapy for prostate cancer.


The Journal of Steroid Biochemistry and Molecular Biology | 1999

Development of (p-O-sulfamoyl)-N-alkanoyl-phenylalkyl amines as non-steroidal estrone sulfatase inhibitors.

Aparna Kolli; Guo-Hua Chu; Michael E. Rhodes; Kengo Inoue; Kyle W. Selcer; Pui-Kai Li

Estrogen levels in breast tumors of postmenopausal women are as much as 10 times higher than estrogen levels in plasma, presumably due to in situ formation of estrogen. The major source of estrogen in breast cancer cells may be conversion of estrone sulfate to estrone by the enzyme estrone sulfatase. Thus, inhibitors of estrone sulfatase are potential agents for treatment of estrogen-dependent breast cancer. Several steroidal compounds have been developed that are potent estrone sulfatase inhibitors, most notably estrone-3-O-sulfamate. However, these compounds and their metabolites may have undesired effects, including estrogenicity. To avoid the problems associated with a potentially active steroid nucleus, we designed and synthesized a series of nonsteroidal estrone sulfatase inhibitors, the (p-O-sulfamoyl)-N-alkanoyl phenylalkyl amines. The compounds synthesized vary in the length of their alkanoyl chain and in the number of carbons separating the phenyl ring and the carbonyl carbon. The ability of these compounds to inhibit estrone sulfatase activity was tested using human placental microsomes and intact cultured human breast cancer cells. Estrogenicity was also evaluated, using growth of estrogen-dependent human breast cancer cells. All of the test compounds inhibited estrone sulfatase activity of human placental microsomes to some extent, with the most effective compound having an IC50 value of 72 nM. In general, compounds with longer alkanoyl chains (12-14 carbons) were more effective than those with shorter chains. The test compounds also inhibited estrone sulfatase activity in intact cultures of MDA-MB-231 human breast cancer cells. Again, the longer chain compounds were more effective. In both the placental and breast cancer cell sulfatase assays, the optimal distance between the phenyl ring and the carbonyl carbon was 1-2 carbons. The MCF-7 cell proliferation assay revealed that estrone and estrone-3-O-sulfamate were both estrogenic, but the (p-O-sulfamoyl)-N-alkanoyl phenylalkyl amines were not. Our data indicate the utility of (p-O-sulfamoyl)-N-alkanoyl phenyl alkylamines for inhibition of estrone sulfatase activity. Furthermore, our data support the concept that nonsteroidal estrone sulfatase inhibitors may be useful as therapeutic agents for estrogen-dependent breast cancers.


The Journal of Steroid Biochemistry and Molecular Biology | 2007

Immunohistochemical analysis of steroid sulfatase in human tissues

Kyle W. Selcer; Heidi DiFrancesca; Abhinav B. Chandra; Pui-Kai Li

Steroid sulfatase (EC 3.1.6.2) is an enzyme that removes the sulfate group from 3beta-hydroxysteroid sulfates. This enzyme is best known for its role in estrogen production via the fetal adrenal-placental pathway during pregnancy; however, it also has important functions in other physiological and pathological steroid pathways. The objective of this study was to examine the distribution of steroid sulfatase in normal human tissues and in breast cancers using immunohistochemistry, employing a newly developed steroid sulfatase antibody. A rabbit polyclonal antiserum was generated against a peptide representing a conserved region of the steroid sulfatase protein. In Western blotting experiments using human placental microsomes, this antiserum crossreacted with a 65 kDa protein, the reported size of steroid sulfatase. The antiserum also crossreacted with single protein bands in Western blots of microsomes from two human breast cancer cell lines (MDA-MB-231 and MCF-7) and from rat liver; however, there were some size differences in the immunoreactive bands among tissues. The steroid sulfatase antibody was used in immunohistochemical analyses of individual human tissue slides as well as a human tissue microarray. For single tissues, human placenta and liver showed strong positive staining against the steroid sulfatase antibody. ER+/PR+ breast cancers also showed relatively strong levels of steroid sulfatase immunoreactivity. Normal human breast showed moderate levels of steroid sulfatase immunoreactivity, while ER-/PR- breast cancer showed weak immunoreactivity. This confirms previous reports that steroid sulfatase is higher in hormone-dependent breast cancers. For the tissue microarray, most tissues showed some detectable level of steroid sulfatase immunoreactivity, but there were considerable differences among tissues, with skin, liver and lymph nodes having the highest immunoreactivity and brain tissues having the lowest. These data reveal the utility of immunohistochemistry in evaluation of steroid sulfatase activity among tissues. The newly developed antibody should be useful in studies of both humans and rats.


Bioorganic & Medicinal Chemistry Letters | 1999

Synthesis and sulfatase inhibitory activities of (E)- and (Z)-4-hydroxytamoxifen sulfamates

Guo-Hua Chu; Amy Peters; Kyle W. Selcer; Pui-Kai Li

We report the development of (E)- and (Z)-4-hydroxytamoxifen sulfamates as estrone sulfatase inhibitors, potential therapeutic agents for the treatment of breast cancer. Both compounds competitively inhibit estrone sulfatase isolated from rat liver with apparent Ki of 35.9 microM for (E)-4-hydroxytamoxifen sulfamate and an apparent Ki of > 500 microM for the (Z) isomer.


Journal of Hepatology | 2016

Inflammatory regulation of steroid sulfatase: A novel mechanism to control estrogen homeostasis and inflammation in chronic liver disease

Mengxi Jiang; Marcus Klein; Ulrich M. Zanger; Mohammad K. Mohammad; Matthew C. Cave; Nilesh W. Gaikwad; Natasha J. Dias; Kyle W. Selcer; Yan Guo; Jinhan He; Xiuhui Zhang; Qiujin Shen; Wenxin Qin; Jiang Li; Song Li; Wen Xie

BACKGROUND & AIMS Chronic inflammatory liver diseases are associated with estrogen excess and feminization in men, which is thought to be due to compromised liver function to break down estrogens. The goal of this study is to determine whether the inflammatory induction of steroid sulfatase (STS), which converts inactive estrogen sulfates to active estrogens, may have contributed to the estrogen excess in chronic liver disease. METHODS We performed bioinformatic analysis, real-time PCR, immunohistochemistry, and UPLC/MS-MS to analyze hepatic STS expression and serum estrogen levels in patients with chronic liver diseases. The crosstalk between NF-κB pathway and STS-regulated estrogen signaling was investigated by electrophoretic mobility shift assay, chromatin immunoprecipitation, luciferase assay and gene knockdown experiments in human hepatocytes. RESULTS Hepatic STS was induced in patients with chronic inflammatory liver diseases, which was accompanied by increased circulating estrogen levels. The human STS gene, but not the mouse Sts gene, was induced by inflammatory stimuli in hepatic cells. Mechanistically, STS was established as a novel NF-κB target gene, whose induction facilitated the conversion of inactive estrogen sulfates to active estrogens, and consequently attenuated the inflammatory response. In contrast, genetic or pharmacological inhibition of STS or a direct blockade of estrogen signaling sensitized liver cells to the transcriptional activation of NF-κB and inflammatory response, possibly through the inhibition of IκB kinase activation. CONCLUSIONS Our results suggest a negative feedback loop in chronic inflammatory liver diseases, in which the inflammatory activation of NF-κB induces STS gene expression. The induced STS facilitates the conversion of inactive estrogen sulfates to active estrogens, which in return attenuates the NF-κB-mediated inflammation.


Journal of Biological Chemistry | 2014

Hepatic Overexpression of Steroid Sulfatase Ameliorates Mouse Models of Obesity and Type 2 Diabetes through Sex-specific Mechanisms

Mengxi Jiang; Jinhan He; Heidi R. Kucera; Nilesh W. Gaikwad; Bin Zhang; Meishu Xu; Robert M. O'Doherty; Kyle W. Selcer; Wen Xie

Background: Steroid sulfatase (STS)-mediated desulfation regulates the chemical and functional homeostasis of estrogens. Results: Overexpression of STS in the liver improved metabolic functions in mouse models of obesity and type 2 diabetes through sex-specific mechanisms. Conclusion: STS-mediated estrogen reactivation is beneficial in energy and glucose metabolism. Significance: Liver-specific activation of estrogen signaling may represent a novel approach to manage metabolic syndrome. The steroid sulfatase (STS)-mediated desulfation is a critical metabolic mechanism that regulates the chemical and functional homeostasis of endogenous and exogenous molecules. In this report, we first showed that the liver expression of Sts was induced in both the high fat diet (HFD) and ob/ob models of obesity and type 2 diabetes and during the fed to fasting transition. In defining the functional relevance of STS induction in metabolic disease, we showed that overexpression of STS in the liver of transgenic mice alleviated HFD and ob/ob models of obesity and type 2 diabetes, including reduced body weight, improved insulin sensitivity, and decreased hepatic steatosis and inflammation. Interestingly, STS exerted its metabolic benefit through sex-specific mechanisms. In female mice, STS may have increased hepatic estrogen activity by converting biologically inactive estrogen sulfates to active estrogens and consequently improved the metabolic functions, whereas ovariectomy abolished this protective effect. In contrast, the metabolic benefit of STS in males may have been accounted for by the male-specific decrease of inflammation in white adipose tissue and skeletal muscle as well as a pattern of skeletal muscle gene expression that favors energy expenditure. The metabolic benefit in male STS transgenic mice was retained after castration. Treatment with the STS substrate estrone sulfate also improved metabolic functions in both the HFD and ob/ob models. Our results have uncovered a novel function of STS in energy metabolism and type 2 diabetes. Liver-specific STS induction or estrogen/estrogen sulfate delivery may represent a novel approach to manage metabolic syndrome.


Copeia | 1988

Effects of pentastome infection on reproduction in a southern Texas population of the Mediterranean Gecko, Hemidactylus turcicus

Danny B. Pence; Kyle W. Selcer

Forty-three percent of adult Mediterranean geckos, Hemidactylus turcicus, from southern Texas were infected with a pulmonary pentastome, Raillietiella frenatus. Pentastome occurrence and biomass did not affect carcass, liver or testis mass of males or carcass mass of females through the year. However, liver mass in females during the nonreproductive period and fatbody mass in both sexes during the reproductive period was significantly lower in individuals with the highest biomass of pentastomes. Infected females had a higher frequency of medium sized yolked follicles than uninfected females, but the latter had a significantly higher frequency of oviductal eggs than infected female geckos. We estimated that oviductal egg production in this gecko population was reduced by 21% due to the pentastome infection. These data indicated that this macroparasitic infection should be considered as a possible factor in the population regulation of H. turcicus in southern Texas.

Collaboration


Dive into the Kyle W. Selcer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mengxi Jiang

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roldán A. Valverde

Southeastern Louisiana University

View shared research outputs
Top Co-Authors

Avatar

Wen Xie

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Wendell W. Leavitt

Worcester Foundation for Biomedical Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge