Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
J Med Chem ; 53(8): 3296-304, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20350005

ABSTRACT

A series of phenyl sulfone substituted quinoxaline were prepared and the lead compound 13 (WYE-672) was shown to be a tissue selective LXR Agonist. Compound 13 demonstrated partial agonism for LXRbeta in kidney HEK-293 cells but did not activate Gal4 LXRbeta fusion proteins in huh-7 liver cells. Although 13 showed potent binding affinity to LXRbeta (IC(50) = 53 nM), it had little binding affinity for LXRalpha (IC(50) > 1.0 microM) and did not recruit any coactivator/corepressor peptides in the LXRalpha multiplex assay. However, compound 13 showed good agonism in THP-1 cells with respect to increasing ABCA1 gene expression and good potency on cholesterol efflux in THP-1 foam cells. In an eight-week lesion study in LDLR -/- mice, compound 13 showed reduction of aortic arch lesion progression and no plasma or hepatic triglyceride increase. These results suggest quinoxaline 13 may have an improved biological profile for potential use as a therapeutic agent.


Subject(s)
Orphan Nuclear Receptors/agonists , Quinoxalines/chemical synthesis , Sulfones/chemical synthesis , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/biosynthesis , Animals , Area Under Curve , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cell Line , Cholesterol/metabolism , Duodenum/metabolism , Half-Life , Humans , Kidney/metabolism , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Organ Specificity , Orphan Nuclear Receptors/genetics , Quinoxalines/chemistry , Quinoxalines/pharmacology , Radioligand Assay , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology , Transcriptional Activation , Triglycerides/metabolism
2.
J Med Chem ; 53(4): 1774-87, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20095622

ABSTRACT

In an effort to develop orally active farnesoid X receptor (FXR) agonists, a series of tetrahydroazepinoindoles with appended solubilizing amine functionalities were synthesized. The crystal structure of the previously disclosed FXR agonist, 1 (FXR-450), aided in the design of compounds with tethered solubilizing functionalities designed to reach the solvent cavity around the hFXR receptor. These compounds were soluble in 0.5% methylcellulose/2% Tween-80 in water (MC/T) for oral administration. In vitro and in vivo optimization led to the identification of 14dd and 14cc, which in a dose-dependent fashion regulated low density lipoprotein cholesterol (LDLc) in low density lipoprotein receptor knockout (LDLR(-/-)) mice. Compound 14cc was dosed in female rhesus monkeys for 4 weeks at 60 mg/kg daily in MC/T vehicle. After 7 days, triglyceride (TG) levels and very low density lipoprotein cholesterol (VLDLc) levels were significantly decreased and LDLc was decreased 63%. These data are the first to demonstrate the dramatic lowering of serum LDLc levels by a FXR agonist in primates and supports the potential utility of 14cc in treating dyslipidemia in humans beyond just TG lowering.


Subject(s)
Azepines/chemical synthesis , Hypolipidemic Agents/chemical synthesis , Indoles/chemical synthesis , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Azepines/pharmacokinetics , Azepines/pharmacology , Biological Availability , Cell Line , Cholesterol, LDL/blood , Female , Humans , Hypolipidemic Agents/pharmacokinetics , Hypolipidemic Agents/pharmacology , Indoles/pharmacokinetics , Indoles/pharmacology , Macaca mulatta , Male , Mice , Mice, Knockout , Microsomes, Liver/metabolism , Models, Molecular , Rats , Rats, Sprague-Dawley , Receptors, LDL/genetics , Solubility , Structure-Activity Relationship , Triglycerides/blood
3.
In Vivo ; 23(5): 827-33, 2009.
Article in English | MEDLINE | ID: mdl-19779119

ABSTRACT

BACKGROUND: The purpose of this study was to investigate the immunological impact of combining neoadjuvant total androgen suppression (TAS) with radiotherapy (xRT) in the treatment of prostate cancer by monitoring blood cytokine levels. PATIENTS AND METHODS: Participants were stage I-II prostate cancer patients receiving xRT alone (n=18) or TAS+xRT (n=19) under the procedures outlined in RTOG protocols #94-08 and #94-13. Peripheral blood samples were collected immediately prior to TAS (xRT+TAS group), immediately prior to xRT, 24 hours after initiation of xRT, and weekly during xRT. Samples were monitored for the immunoregulatory cytokines interleukin (IL)-1beta, IL-6 and transforming growth factor (TGF)beta using ELISA procedures. RESULTS: Following initiation of xRT, both patient groups demonstrated an immediate elevation of the proinflammatory cytokines IL-1beta and IL-6 in their plasma. These cytokine levels appeared to peak after 1-2 weeks of xRT before returning toward pre xRT levels. In contrast, the profibrotic cytokine TGFbeta appeared to decrease immediately following initiation of xRT, but, subsequently, underwent two distinct waves of elevation, occurring at 1-2 weeks and 5-6 weeks into the xRT. Surprisingly, while the temporal pattern of plasma cytokine response was similar in both treatment groups, the magnitude of cytokine expression was noticeably different, appearing to be significantly affected by the addition of TAS. Indeed, administration of neoadjuvant TAS appeared to bring about a marked elevation of IL-1beta and IL-6 and a significant reduction in TGFbeta when compared to patients receiving xRT alone. CONCLUSION: The precise mechanisms underlying this TAS-related increase of the proinflammatory cytokines IL-1beta and IL-6 and decrease of the profibrotic cytokine TGFbeta remain unclear. However, previous reports have documented that androgens tend to be immunosuppressive in nature. It is conceivable, therefore, that administration of TAS shifts the ratio of proinflammatory and profibrotic cytokines toward a more immunostimulatory state.


Subject(s)
Adenocarcinoma/blood , Androgen Antagonists/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Cytokines/blood , Prostatic Neoplasms/blood , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Aged , Humans , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Radiotherapy, Adjuvant
4.
Bioorg Med Chem Lett ; 19(18): 5289-92, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19683924

ABSTRACT

Pyrrole[2,3-d]azepines have been identified as potent agonists of the farnesoid X receptor (FXR). Based on the planar X-ray crystal structure of WAY-362450 1 in the ligand binding domain and molecular modeling studies, non-planar reduced compounds were designed which led to agonists that exhibit high aqueous solubility and retain moderate in vitro potency.


Subject(s)
Azepines/pharmacology , Pyrroles/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Azepines/chemistry , Humans , Models, Molecular , Protein Binding , Pyrroles/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Structure-Activity Relationship
5.
J Lipid Res ; 50(12): 2358-70, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19318684

ABSTRACT

Liver X receptors (LXRs) are ligand-activated transcription factors that coordinate regulation of gene expression involved in several cellular functions but most notably cholesterol homeostasis encompassing cholesterol transport, catabolism, and absorption. WAY-252623 (LXR-623) is a highly selective and orally bioavailable synthetic modulator of LXR, which demonstrated efficacy for reducing lesion progression in the murine LDLR(-/-) atherosclerosis model with no associated increase in hepatic lipogenesis either in this model or Syrian hamsters. In nonhuman primates with normal lipid levels, WAY-252623 significantly reduced total (50-55%) and LDL-cholesterol (LDLc) (70-77%) in a time- and dose-dependent manner as well as increased expression of the target genes ABCA1/G1 in peripheral blood cells. Statistically significant decreases in LDLc were noted as early as day 7, reached a maximum by day 28, and exceeded reductions observed for simvastatin alone (20 mg/kg). Transient increases in circulating triglycerides and liver enzymes reverted to baseline levels over the course of the study. Complementary microarray analysis of duodenum and liver gene expression revealed differential activation of LXR target genes and suggested no direct activation of hepatic lipogenesis. WAY-252623 displays a unique and favorable pharmacological profile suggesting synthetic LXR ligands with these characteristics may be suitable for evaluation in patients with atherosclerotic dyslipidemia.


Subject(s)
Atherosclerosis/drug therapy , Cholesterol, LDL/drug effects , Cholesterol, LDL/metabolism , Indazoles/pharmacology , Lipid Metabolism/drug effects , Macaca fascicularis/metabolism , Orphan Nuclear Receptors/agonists , Animals , Atherosclerosis/metabolism , Caco-2 Cells , Cricetinae , Disease Models, Animal , Humans , Indazoles/blood , Indazoles/chemistry , Ligands , Liver/enzymology , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors/metabolism
6.
J Lipid Res ; 50(6): 1090-100, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19174369

ABSTRACT

The role of farnesoid X receptor (FXR) in the development of atherosclerosis has been unclear. Here, LDL receptor (LDLR(-/-)) or apolipoprotein E (apoE(-/-)) female or male mice were fed a Western diet and treated with a potent synthetic FXR agonist, WAY-362450. Activation of FXR blocked diet-induced hypertriglyceridemia and elevations of non-HDL cholesterol and produced a near complete inhibition of aortic lesion formation. WAY-362450 also induced small heterodimer partner (SHP) expression and repressed cholesterol 7alpha-hydroxylase (CYP7A1) and sterol 12 alpha-hydroxylase (CYP8B1) expression. To determine if SHP was essential for these protective activities, LDLR(-/-)SHP(-/-) and apoE(-/-)SHP(-/-) mice were similarly treated with WAY-362450. Surprisingly, a notable sex difference was observed in these mice. In male LDLR(-/-)SHP(-/-) or apoE(-/-)SHP(-/-) mice, WAY-362450 still repressed CYP7A1 and CYP8B1 expression by 10-fold and still strongly reduced non-HDL cholesterol levels and aortic lesion area. In contrast, in the female LDLR(-/-)SHP(-/-) or apoE(-/-)SHP(-/-) mice, WAY-362450 only slightly repressed CYP7A1 and CYP8B1 expression and did not reduce non-HDL cholesterol or aortic lesion size. WAY-362450 inhibition of hypertriglyceridemia remained intact in LDLR(-/-) or apoE(-/-) mice lacking SHP of both sexes. These results suggest that activation of FXR protects against atherosclerosis in the mouse, and this protective effect correlates with repression of bile acid synthetic genes, with mechanistic differences between male and female mice.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, LDL/deficiency , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Azepines/pharmacology , Bile Acids and Salts/metabolism , Cholesterol 7-alpha-Hydroxylase/antagonists & inhibitors , Cholesterol 7-alpha-Hydroxylase/genetics , Dyslipidemias/genetics , Dyslipidemias/metabolism , Dyslipidemias/prevention & control , Female , Gene Expression/drug effects , Indoles/pharmacology , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, LDL/genetics , Steroid 12-alpha-Hydroxylase/antagonists & inhibitors , Steroid 12-alpha-Hydroxylase/genetics
7.
Am J Physiol Gastrointest Liver Physiol ; 296(3): G543-52, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19136377

ABSTRACT

The nuclear hormone receptor farnesoid X receptor (FXR) plays a critical role in the regulation of bile acid, triglyceride (TG), and cholesterol homeostasis. WAY-362450 (FXR-450/XL335) is a potent synthetic FXR agonist as characterized in luciferase reporter assays and in mediating FXR target gene regulation in primary human and immortalized mouse hepatocytes. In vivo, WAY-362450 dose dependently decreased serum TG levels after 7 days of oral dosing in western diet-fed low-density lipoprotein receptor-/- mice and in the diabetic mouse strains KK-Ay and db/db comparable to that achieved with the peroxisome proliferator activated receptor-alpha agonist, fenofibrate. WAY-362450 treatment also reduced serum cholesterol levels via reductions in LDLc, VLDLc, and HDLc lipoprotein fractions that were not accompanied by hepatic cholesterol accumulation. This cholesterol lowering was dependent on FXR as demonstrated in a hypothyroid-induced hypercholesterolemia setting in FXR-/- mice. In fructose-fed models, WAY-362450 also decreased TG and VLDLc levels in rats and hamsters but significantly increased HDLc levels in rats while reducing HDLc levels in hamsters. The differential effect of WAY-362450 on HDLc is likely due to a murine-specific induction of endothelial lipase and scavenger receptor-BI that does not occur in rats. These studies demonstrate a consistent ability of WAY-362450 to lower both serum TG and cholesterol levels and suggest that synthetic FXR agonists may have clinical utility in the treatment of mixed dyslipidemia.


Subject(s)
Azepines/pharmacology , Cholesterol/blood , DNA-Binding Proteins/agonists , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Indoles/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Animals , Apolipoprotein A-I/blood , Apolipoprotein A-I/genetics , Azepines/chemistry , Cells, Cultured , Cholesterol/pharmacology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Dyslipidemias/complications , Female , Fructose/pharmacology , Humans , Hyperglycemia/complications , Hyperglycemia/genetics , Hyperinsulinism/complications , Hyperinsulinism/genetics , Indoles/chemistry , Kidney/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, LDL/genetics , Receptors, Leptin/genetics , Transcription Factors/metabolism , Triglycerides/blood
8.
J Lipid Res ; 50(2): 193-203, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18820241

ABSTRACT

Multiple studies suggest increased conversion of cholesterol to bile acids by cholesterol 7alpha-hydroxylase (CYP7A1) protects against dyslipidemia and atherosclerosis. CYP7A1 expression is repressed by the sequential activity of two nuclear hormone receptors, farnesoid X receptor (FXR) and small heterodimer partner (SHP). Here we demonstrate 129 strain SHP(-/-) mice are protected against hypercholesterolemia resulting from either a cholesterol/cholic acid (chol/CA) diet or from hypothyroidism. In a mixed 129-C57Bl/6 background, LDLR(-/-) and LDLR(-/-)SHP(-/-) mice had nearly identical elevations in hepatic cholesterol content and repression of cholesterol regulated genes when fed a Western diet. However, the LDLR(-/-)SHP(-/-) mice had greatly reduced elevations in serum VLDL and LDL cholesterol levels and triglyceride (TG) levels as compared with LDLR(-/-) mice. Additionally, the hepatic inflammation produced by the Western diet in the LDLR(-/-) mice was abolished in the LDLR(-/-)SHP(-/-) mice. CYP7A1 expression was induced 10-fold by the Western diet in the LDLR(-/-)SHP(-/-) mice but not in the LDLR(-/-) mice. Finally, hepatocyte-specific deletion of SHP expression was also protective against dyslipidemia induced by either a chol/CA diet or by hypothyroidism. While no antagonist ligands have yet been identified for SHP, these results suggest selective inhibition of hepatic SHP expression may provide protection against dyslipidemia.


Subject(s)
Dyslipidemias/genetics , Dyslipidemias/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Cholesterol, LDL/blood , Cholesterol, LDL/metabolism , Cholesterol, VLDL/blood , Cholesterol, VLDL/metabolism , Gene Expression , Mice , Mice, Transgenic , Receptors, Cytoplasmic and Nuclear/metabolism
9.
J Med Chem ; 51(22): 7161-8, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18973288

ABSTRACT

A series of substituted 2-benzyl-3-aryl-7-trifluoromethylindazoles were prepared as LXR modulators. These compounds were partial agonists in transactivation assays when compared to 1 (T0901317) and were slightly weaker with respect to potency and efficacy on LXRalpha than on LXRbeta. Lead compounds in this series 12 (WAY-252623) and 13 (WAY-214950) showed less lipid accumulation in HepG2 cells than potent full agonists 1 and 3 (WAY-254011) but were comparable in efficacy to 1 and 3 with respect to cholesterol efflux in THP-1 foam cells, albeit weaker in potency. Compound 13 reduced aortic lesion area in LDLR knockout mice equivalently to 3 or positive control 2 (GW3965). In a 7-day hamster model, compound 13 showed a lesser propensity for plasma TG elevation than 3, when the compounds were compared at doses in which they elevated ABCA1 and ABCG1 gene expression in duodenum and liver at equal levels. In contrast to results previously published for 2, the lack of TG effect of 13 correlated with its inability to increase liver fatty acid synthase (FAS) gene expression, which was up-regulated 4-fold by 3. These results suggest indazoles such as 13 may have an improved profile for potential use as a therapeutic agent.


Subject(s)
Arteriosclerosis/drug therapy , DNA-Binding Proteins/agonists , Indazoles/pharmacology , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Triglycerides/biosynthesis , Animals , Arteriosclerosis/metabolism , Cell Differentiation/drug effects , Cell Line , Cricetinae , Crystallography, X-Ray , DNA-Binding Proteins/metabolism , Humans , Hydrogen Bonding , Indazoles/chemical synthesis , Indazoles/chemistry , Ligands , Liver/drug effects , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Models, Molecular , Molecular Structure , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Structure-Activity Relationship , Triglycerides/blood
10.
J Transl Med ; 6: 59, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18925943

ABSTRACT

BACKGROUND: LXRs (Liver X Receptor alpha and beta) are nuclear receptors that act as ligand-activated transcription factors. LXR activation causes upregulation of genes involved in reverse cholesterol transport (RCT), including ABCA1 and ABCG1 transporters, in macrophage and intestine. Anti-atherosclerotic effects of synthetic LXR agonists in murine models suggest clinical utility for such compounds. OBJECTIVE: Blood markers of LXR agonist exposure/activity were sought to support clinical development of novel synthetic LXR modulators. METHODS: Transcript levels of LXR target genes ABCA1 and ABCG1 were measured using quantitative reverse transcriptase/polymerase chain reaction assays (qRT-PCR) in peripheral blood from mice and rats (following a single oral dose) and monkeys (following 7 daily oral doses) of synthetic LXR agonists. LXRalpha, LXRbeta, ABCA1, and ABCG1 mRNA were measured by qRT-PCR in human peripheral blood mononuclear cells (PBMC), monocytes, T- and B-cells treated ex vivo with WAY-252623 (LXR-623), and protein levels in human PBMC were measured by Western blotting. ABCA1/G1 transcript levels in whole-blood RNA were measured using analytically validated assays in human subjects participating in a Phase 1 SAD (Single Ascending Dose) clinical study of LXR-623. RESULTS: A single oral dose of LXR agonists induced ABCA1 and ABCG1 transcription in rodent peripheral blood in a dose- and time-dependent manner. Induction of gene expression in rat peripheral blood correlated with spleen expression, suggesting LXR gene regulation in blood has the potential to function as a marker of tissue gene regulation. Transcriptional response to LXR agonist was confirmed in primates, where peripheral blood ABCA1 and ABCG1 levels increased in a dose-dependent manner following oral treatment with LXR-623. Human PBMC, monocytes, T- and B cells all expressed both LXRalpha and LXRbeta, and all cell types significantly increased ABCA1 and ABCG1 expression upon ex vivo LXR-623 treatment. Peripheral blood from a representative human subject receiving a single oral dose of LXR-623 showed significant time-dependent increases in ABCA1 and ABCG1 transcription. CONCLUSION: Peripheral blood cells express LXRalpha and LXRbeta, and respond to LXR agonist treatment by time- and dose-dependently inducing LXR target genes. Transcript levels of LXR target genes in peripheral blood are relevant and useful biological indicators for clinical development of synthetic LXR modulators.


Subject(s)
Blood Cells/metabolism , DNA-Binding Proteins/agonists , Receptors, Cytoplasmic and Nuclear/agonists , Transcription, Genetic , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Administration, Oral , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/pharmacology , Biomarkers , Blood Cells/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Liver X Receptors , Orphan Nuclear Receptors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
11.
Anticancer Res ; 28(6A): 3601-7, 2008.
Article in English | MEDLINE | ID: mdl-19189640

ABSTRACT

BACKGROUND: Interleukin-1alpha (IL-1) is known to radioprotect the gastrointestinal tract, but the mechanism by which this protection occurs remains unclear. These studies were undertaken to investigate whether the radioprotective potential of IL-1 may be linked to an ability to reduce apoptosis within the gastrointestinal crypts. MATERIALS AND METHODS: IL-1 was administered to C57Bl/6 mice 24 hours prior to receiving 8 Gy abdominal X-irradiation (xRT). At designated times, experimental mice were sacrificed, jejunal tissue removed, and paraffin-embedded sections analyzed for apoptosis indices (AI) and immunohistochemical determination of active caspase-3, -8 and -9. RESULTS: AI data demonstrated that 8 Gy irradiation resulted in a marked jejunal apoptotic response, but IL-1 pretreatment significantly attenuated this response. Concomitant with this attenuation, reduced levels of caspase-3 and 9, but not caspase-8, activation were observed, particularly within goblet cells. CONCLUSION: The results outlined herein suggest that radioprotection by IL-1 is mediated, at least in part, through a reduction in the apoptotic response which appears to involve down-regulation of the intrinsic apoptotic pathway.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Interleukin-1alpha/pharmacology , Jejunum/drug effects , Jejunum/radiation effects , Radiation-Protective Agents/pharmacology , Animals , Caspases/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/radiation effects , Enzyme Activation/radiation effects , Female , In Vitro Techniques , Isoenzymes/metabolism , Jejunum/enzymology , Jejunum/pathology , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control
12.
Cancer Biol Ther ; 5(8): 1022-30, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16855386

ABSTRACT

Many cancer treatments induce cell death through lethal oxidative stress. Oxidative stress also induces the activation of the calcium/calmodulin-dependent kinases (CaM-Ks), CaM-KII and CaM-KIV. In turn, the CaM-Ks are known to induce the activation of antiapoptotic signaling pathways, such as Akt, ERK, and NF-kappaB in many different cell types. The aim of this study was to determine the role of CaM-Kinases in resistance to hydrogen peroxide and three oxidative stress-inducing cancer therapies in MCF-7 breast cancer cells. We found that oxidative stress induced CaM-Kinase activity in MCF-7 breast cancer cells and that CaM-K inhibition increased hydrogen peroxide-induced cell death in MCF-7 human breast cancer cells. When MCF-7 cells were treated with doxorubicin, ionizing radiation, or photodynamic therapy in the presence of a CaM-K inhibitor a greater level of cell killing was observed than when cells were treated with doxorubicin, ionizing radiation, or photodynamic therapy alone. In support of this finding, CaM-K inhibition increased hydrogen peroxide-induced apoptosis in MCF-7 cells, as determined by increased number of apoptotic cells, DNA fragmentation, and PARP cleavage. Pharmacological and molecular inhibition indicated that CaM-KII was participating in hydrogen peroxide-induced ERK phosphorylation in breast cancer cells indicating a potential mechanism by which this sensitization occurs. This is the first time that CaM-K inhibition is reported to sensitize cancer cells to reactive oxygen intermediate inducing cancer treatments.


Subject(s)
Apoptosis , Breast Neoplasms/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Reactive Oxygen Species/pharmacology , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinase Type 4 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Doxorubicin/pharmacology , Flow Cytometry , Humans , Hydrogen Peroxide/pharmacology , Immunoblotting , Mitogen-Activated Protein Kinase 3/metabolism , Oxidants/pharmacology , Oxidative Stress , Phosphorylation/drug effects , Photochemotherapy , RNA, Small Interfering/pharmacology , Radiation, Ionizing , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
13.
Endocrinology ; 147(9): 4025-33, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16675527

ABSTRACT

Farnesoid X receptor (FXR) uses bile acids as endogenous ligands. Here, we demonstrate that androsterone, a metabolic product of testosterone, is also an FXR ligand. Treatment of castrated male mice with androsterone induced expression of the FXR target gene small heterodimer partner (SHP). In mouse AML-12 hepatocytes, chenodeoxycholic acid (CDCA) or androsterone induced SHP expression with a similar kinetic pattern. The FXR antagonist guggulsterone blocked the induction of SHP by androsterone in AML-12 cells. Nuclear magnetic resonance spectroscopy demonstrated the direct binding of androsterone to purified human FXR (hFXR) ligand-binding domain (LBD) protein, resulting in the recruitment of steroid receptor coactivator protein-1 (SRC-1) coactivator peptide. In HEK293 cells, androsterone activated gal4-mouse FXR-LBD and gal4-hFXR-LBD fusion proteins, although in contrast to CDCA, androsterone activation was significantly greater for the mouse FXR-LBD than for the hFXR-LBD. Site-directed mutagenesis of the hFXR-LBD defined amino acids Asn354 and Ser345 as critical for differential species sensitivity to CDCA and androsterone, respectively. Crystal structure studies suggest that the orientation of the steroid nucleus of bile acids within the binding pocket of FXR is reversed from all other nuclear hormone receptors. In support of this model, we show here that mutations M265I or R331H, residues predicted by crystal structure to interact with the carboxylic acid tail of CDCA but not with androsterone, altered CDCA activation but had no effect on androsterone activation. Activation of FXR by androsterone may provide an additional means for physiological or pharmacological modulation of FXR.


Subject(s)
Androsterone/pharmacology , DNA-Binding Proteins/physiology , Transcription Factors/physiology , Amino Acid Sequence , Androsterone/metabolism , Animals , Cell Line , Chenodeoxycholic Acid/pharmacology , Crystallization , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Embryo, Mammalian , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Histone Acetyltransferases , Humans , Kidney , Ligands , Magnetic Resonance Spectroscopy , Male , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Receptor Coactivator 1 , Orchiectomy , Pregnenediones/pharmacology , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Fusion Proteins , Saccharomyces cerevisiae Proteins/genetics , Structure-Activity Relationship , Testosterone/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Anticancer Res ; 25(4): 3159-66, 2005.
Article in English | MEDLINE | ID: mdl-16080580

ABSTRACT

BACKGROUND: This study sought to better define the immunological impact of combining neoadjuvant total androgen suppression (TAS) with radiotherapy (xRT) in treating prostate cancer. MATERIALS AND METHODS: Subjects selected (n = 37) were stage I-II prostate cancer patients meeting the eligibility requirements for RTOG protocols 94-08 or 94-13. Flow cytometric monitoring of circulating T helper (Th), T suppressor/cytotoxic (Ts), natural killer (NK) and B lymphocytes was performed weekly. RESULTS: Significant reduction of all lymphocyte subsets occurred as a result of xRT. Comparison between treatment groups demonstrated that the B lymphocyte and NK lymphocyte radioresponse was not influenced by TAS, but the Th and Ts lymphocyte response was, with addition of TAS leading to less radiation-induced decline. CONCLUSION: The basis for this T cell response is unclear, but may involve a TAS-induced reduction of testosterone's immunomodulation of T cell proliferation and apoptosis and/or a direct, TAS-induced thymic stimulation. Our data suggest that addition of TAS to xRT appears to have no detrimental effects on lymphocyte subsets, and, indeed, may have favorable effects on T cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , T-Lymphocytes/immunology , Aged , Androgen Antagonists/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Flutamide/administration & dosage , Goserelin/administration & dosage , Humans , Lymphocyte Activation , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , T-Lymphocytes/drug effects , T-Lymphocytes/radiation effects
15.
Am J Physiol Gastrointest Liver Physiol ; 289(2): G267-73, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15817812

ABSTRACT

Previous studies have demonstrated a dramatic induction of inflammatory gene expression in livers from mice fed a high-fat, high-cholesterol diet containing cholate after 3-5 wk. To determine the contribution of cholate in mediating these inductions, C57BL/6 mice were fed a chow diet supplemented with increasing concentrations of cholic acid (CA) for 5 days. A dose-dependent induction in the hepatic levels of TNF-alpha, VCAM-1, ICAM-1, and SAA-2 mRNA were observed. As positive controls, a dose-dependent repression of cholesterol 7alpha-hydroxylase and a dose-dependent induction of small heterodimer partner (SHP) expression were also observed, suggesting that farnesoid X receptor (FXR) was activated. In addition, ICAM-1 and SHP mRNA levels were also induced in primary human hepatocytes when treated with chenodeoxycholic acid or GW4064, a FXR-selective agonist. The involvement of FXR in CA-induced inflammatory gene expression was further investigated in the human hepatic cell line HepG2. Both ICAM-1 and SHP expression were induced in a dose- and time-dependent manner by treatment with the FXR-selective agonist GW4064. Moreover, the induction of ICAM-1 by GW4064 was inhibited by the FXR antagonist guggulsterone or with transfection of FXR siRNA. Finally, the activity of FXR was mapped to a retinoic acid response element (RARE) site containing an imbedded farnesoid X response element (FXRE) on the human ICAM-1 promoter and FXR and retinoid X receptor were demonstrated to bind to this site. Finally, FXR-mediated activation of ICAM-1 could be further enhanced by TNF-alpha cotreatment in hepatocytes, suggesting a potential cooperation between cytokine and bile acid-signaling pathways during hepatic inflammatory events.


Subject(s)
Cholic Acids/pharmacology , DNA-Binding Proteins/metabolism , Hepatocytes/physiology , Intercellular Adhesion Molecule-1/genetics , Signal Transduction/immunology , Transcription Factors/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/agonists , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Gene Expression/immunology , Hepatocytes/cytology , Humans , Isoxazoles/pharmacology , Liver/cytology , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear , Response Elements/genetics , Signal Transduction/genetics , Transcription Factors/agonists , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/pharmacology
17.
Dig Dis Sci ; 49(11-12): 1889-98, 2004.
Article in English | MEDLINE | ID: mdl-15628722

ABSTRACT

In order to diagnose colon cancer at an earlier, more localized stage, there is a need to develop diagnostic markers (genes) which can detect early patterns of gene expression in exfoliated colonocytes shed in the stool during routine screening for this disease. An RNA-based detection is more pertinent than either a DNA-based or a protein-based method as a screening procedure, but it has not been widely used as a cancer screen because of the difficulty of handling and stabilizing the RNA molecule. We describe a method that permits extraction of intact nondegraded total RNA from human colonocytes in stool and from normal and malignant colon tissues (which were employed for comparison with stool). Because it utilizes commercially available kits, this method is simpler than other published methods and does not require isolation of messenger (m)RNA, thereby reducing the chances of contaminating the preparations with degrading nucleases, and even a small amount of isolated total RNA can be adequately reverse transcribed, making high-quality copy (c) DNA. This is followed by PCR (either qualitative end point or semiquantitative real-time) using colon cancer-specific gene primers. By routinely and systematically being able to perform quantitative gene expression measurements on noninvasive samples, the goal of this pilot work is to lay the groundwork for conducting a large clinical study to identify groups of selected genes whose expression is consistently altered at an early stage in the neoplastic process. Such work will permit noninvasive monitoring of at-risk patients through the analysis of their stool samples. Correct diagnosis will allow for surgical and/or other interventions before the tumor is well established and, thus, should decrease mortality from this preventable disease.


Subject(s)
Colon/cytology , Feces/cytology , Intestinal Mucosa/cytology , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction/methods , Adenocarcinoma/diagnosis , Caco-2 Cells , Colonic Neoplasms/diagnosis , DNA, Single-Stranded , Humans , Reagent Kits, Diagnostic , Reproducibility of Results , Sensitivity and Specificity
18.
Anticancer Res ; 23(3B): 2625-31, 2003.
Article in English | MEDLINE | ID: mdl-12894550

ABSTRACT

The response of the blood-forming and supportive stromal compartments of the marrow to a localized, fractionated course of radiotherapy [FxRT; 2.0 Gy (q24 h x 5)-->74 Gy] was examined in the C57Bl/6 mouse using the hematopoietic progenitor and the cobblestone-forming cell (CAFC) assays as end-points for the blood-forming compartment and the long-term bone marrow culture (LTBMC) to assess stromal integrity. Both during and subsequent to FxRT, hematopoietic activity in the irradiated femur was significantly dampened, although an abortive attempt at recovery was observed subsequent to the completion of FxRT. Moreover, both the CAFC subpopulations as well as the functional integrity of LTBMC generated from the irradiated femur were significantly compromised by FxRT. Of interest, restoration of the more primitive CAFC subpopulations to near normal levels was observed in the absence of a comparable recovery of the more mature CAFC subpopulations and the restoration of stromal integrity to the irradiated marrow. As a result, the data are consistent with the generation of a persisting FxRT-induced lesion in the microenvironmental stroma that effectively interferes with the normal regulation of hematopoietic differentiation and the recovery of hematopoietic activity in the irradiated marrow.


Subject(s)
Bone Marrow Cells/radiation effects , Hematopoietic Stem Cells/radiation effects , Animals , Bone Marrow Cells/cytology , Dose Fractionation, Radiation , Mice , Mice, Inbred C57BL , Stromal Cells/radiation effects
19.
J Biol Chem ; 278(38): 36418-29, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12842887

ABSTRACT

Hormonal status can influence diverse metabolic pathways. Small heterodimer partner (SHP) is an orphan nuclear receptor that can modulate the activity of several transcription factors. Estrogens are here shown to directly induce expression of the SHP in the mouse and rat liver and in human HepG2 cells. SHP is rapidly induced within 2 h following treatment of mice with ethynylestradiol (EE) or the estrogen receptor alpha (ERalpha)-selective compound propyl pyrazole triol (PPT). SHP induction by these estrogens is completely absent in ERalphaKO mice. Mutation of the human SHP promoter defined HNF-3, HNF-4, GATA, and AP-1 sites as important for basal activity, whereas EE induction required two distinct elements located between -309 and -267. One of these elements contains an estrogen response element half-site that bound purified ERalpha, and ERalpha with a mutated DNA binding domain was unable to stimulate SHP promoter activity. This ERalpha binding site overlaps the known farnesoid X receptor (FXR) binding site in the SHP promoter, and the combination of EE plus FXR agonists did not produce an additive induction of SHP expression in mice. Surprisingly, induction of SHP by EE did not inhibit expression of the known SHP target genes cholesterol 7alpha-hydroxylase (CYP7A1) or sterol 12alpha-hydroxylase (CYP8B1). However, the direct regulation of SHP expression may provide a basis for some of the numerous biological effects of estrogens.


Subject(s)
Estradiol/analogs & derivatives , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , Cholesterol 7-alpha-Hydroxylase/chemistry , DNA-Binding Proteins/chemistry , Dimerization , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogen Receptor alpha , Gene Deletion , Humans , Liver/metabolism , Male , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Phenols , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Pyrazoles/pharmacology , RNA/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Steroid 12-alpha-Hydroxylase/chemistry , Steroid 12-alpha-Hydroxylase/metabolism , Time Factors , Transcription Factors/chemistry , Transfection
20.
Endocrinology ; 144(2): 701-10, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538633

ABSTRACT

Estrogen receptors (ERs) are expressed in numerous organs, although only a few organs are considered classical targets for estrogens. We have completed a systematic survey of estrogen regulation of approximately 10,000 genes in 13 tissues from wild-type and ERbetaKO mice treated sc with vehicle or 17beta-estradiol (E2) for 6 wk. The uterus and pituitary had the greatest number of genes regulated by E2, whereas the kidney had the third largest number of regulated genes. In situ hybridizations localized E2 regulation in the kidney to the juxtamedullary region of the cortex in both the mouse and rat. The ED(50) for gene inductions in the kidney was 3 micro g/kg.d, comparable with the 2.4 micro g/kg.d ED(50) for c-fos induction in the uterus. E2 regulations in the kidney were intact in ERbetaKO mice, and the ERalpha-selective agonist propylpyrazole triol acted similarly to E2, together suggesting an ERalpha-mediated mechanism. Several genes were induced within 2 h of E2 treatment, suggesting a direct activity of ERalpha within the kidney. Finally, the combination of the activation function (AF)1-selective agonist tamoxifen plus ERalphaKO(CH) mice expressing an AF1-deleted version of ERalpha allowed delineation of genes with differing requirements for AF1 or AF2 activity in the kidney.


Subject(s)
Estrogens/pharmacology , Gene Expression/drug effects , Kidney/physiology , Receptors, Estrogen/metabolism , Transcription, Genetic/physiology , Animals , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Gene Expression/physiology , Mice , Mice, Inbred Strains , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Pituitary Gland/physiology , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/genetics , Uterus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL