Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell Rep ; 42(12): 113461, 2023 12 26.
Article in English | MEDLINE | ID: mdl-37979170

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype with no targeted therapeutics. The luminal androgen receptor (LAR) subtype constitutes 15% of TNBC and is enriched for androgen receptor (AR) and AR target genes. Here, we show that a cohort of TNBC not only expresses AR at a much higher rate (∼80%) but also expresses AR splice variants (AR-SVs) (∼20%), further subclassifying LAR-TNBC. Higher AR and AR-SV expression and corresponding aggressive phenotypes are observed predominantly in specimens obtained from African American women. LAR TNBC specimens are enriched for interferon, Janus kinase (JAK)-signal activator and transducer (STAT), and androgen signaling pathways, which are exclusive to AR-expressing epithelial cancer cells. AR- and AR-SV-expressing TNBC cell proliferation and xenograft and patient-tumor explant growth are inhibited by AR N-terminal domain-binding selective AR degrader or by a JAK inhibitor. Biochemical analysis suggests that STAT1 is an AR coactivator. Collectively, our work identifies pharmacologically targetable TNBC subtypes and identifies growth-promoting interaction between AR and JAK-STAT signaling.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic
2.
J Med Chem ; 66(5): 3372-3392, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36825758

ABSTRACT

A major challenge for new drug discovery in the area of androgen receptor (AR) antagonists lies in predicting the druggable properties that will enable small molecules to retain their potency and stability during further studies in vitro and in vivo. Indole (compound 8) is a first-in-class AR antagonist with very high potency (IC50 = 0.085 µM) but is metabolically unstable. During the metabolic studies described herein, we synthesized new small molecules that exhibit significantly improved stability while retaining potent antagonistic activity for an AR. This structure-activity relationship (SAR) study of more than 50 compounds classified with three classes (Class I, II, and III) and discovered two compounds (32c and 35i) that are potent AR antagonists (e.g., IC50 = 0.021 µM, T1/2 = 120 min for compound 35i). The new antagonists exhibited improved in vivo pharmacokinetics (PK) with high efficacy antiandrogen activity in Hershberger and antiandrogen Enz-Res tumor xenograft models that overexpress AR (LNCaP-AR).


Subject(s)
Androgen Receptor Antagonists , Prostatic Neoplasms , Male , Humans , Androgen Receptor Antagonists/pharmacology , Androgen Receptor Antagonists/therapeutic use , Receptors, Androgen/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Androgen Antagonists , Nitriles/pharmacology , Cell Line, Tumor , Cell Proliferation
3.
Proc Natl Acad Sci U S A ; 120(1): e2211832120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36577061

ABSTRACT

Androgen receptor (AR) and its splice variants (AR-SVs) promote prostate cancer (PCa) growth by orchestrating transcriptional reprogramming. Mechanisms by which the low complexity and intrinsically disordered primary transactivation domain (AF-1) of AR and AR-SVs regulate transcriptional programming in PCa remains poorly defined. Using omics, live and fixed fluorescent microscopy of cells, and purified AF-1 and AR-V7 recombinant proteins we show here that AF-1 and the AR-V7 splice variant form molecular condensates by liquid-liquid phase separation (LLPS) that exhibit disorder characteristics such as rapid intracellular mobility, coactivator interaction, and euchromatin induction. The LLPS and other disorder characteristics were reversed by a class of small-molecule-selective AR-irreversible covalent antagonists (SARICA) represented herein by UT-143 that covalently and selectively bind to C406 and C327 in the AF-1 region. Interfering with LLPS formation with UT-143 or mutagenesis resulted in chromatin condensation and dissociation of AR-V7 interactome, all culminating in a transcriptionally incompetent complex. Biochemical studies suggest that C327 and C406 in the AF-1 region are critical for condensate formation, AR-V7 function, and UT-143's irreversible AR inhibition. Therapeutically, UT-143 possesses drug-like pharmacokinetics and metabolism properties and inhibits PCa cell proliferation and tumor growth. Our work provides critical information suggesting that clinically important AR-V7 forms transcriptionally competent molecular condensates and covalently engaging C327 and C406 in AF-1, dissolves the condensates, and inhibits its function. The work also identifies a library of AF-1-binding AR and AR-SV-selective covalent inhibitors for the treatment of PCa.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Receptors, Androgen/metabolism , Cysteine , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Androgen Receptor Antagonists/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Cell Line, Tumor , Protein Isoforms/metabolism
4.
J Med Chem ; 64(15): 11045-11062, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34269581

ABSTRACT

A series of propanamide derivatives were designed, synthesized, and pharmacologically characterized as selective androgen receptor degraders (SARDs) and pan-antagonists that exert a broad-scope androgen receptor (AR) antagonism. Incorporating different basic heteromonocyclic B-ring structural elements in the common A-ring-linkage-B-ring nonsteroidal antiandrogen general pharmacophore contributed to a novel scaffold of small molecules with SARD and pan-antagonist activities even compared to our recently published AF-1 binding SARDs such as UT-69 (11), UT-155 (12), and UT-34 (13). Compound 26f exhibited inhibitory and degradation effects in vitro in a wide array of wtAR, point mutant, and truncation mutant-driven prostate cancers (PCs). Further, 26f inhibited tumor cell growth in a xenograft model composed of enzalutamide-resistant (EnzR) LNCaP cells. These results demonstrate an advancement toward the development of novel SARDs and pan-antagonists with efficacy against EnzR prostate cancers.


Subject(s)
Amides/pharmacology , Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Amides/chemical synthesis , Amides/chemistry , Androgen Receptor Antagonists/chemical synthesis , Androgen Receptor Antagonists/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzamides/pharmacology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Male , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nitriles/pharmacology , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
5.
J Med Chem ; 63(21): 12642-12665, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33095584

ABSTRACT

We report herein the design, synthesis, and pharmacological characterization of a library of novel aryl pyrazol-1-yl-propanamides as selective androgen receptor degraders (SARDs) and pan-antagonists that exert broad-scope AR antagonism. Pharmacological evaluation demonstrated that introducing a pyrazole moiety as the B-ring structural element in the common A-ring-linkage-B-ring nonsteroidal antiandrogens' general pharmacophore allowed the development of a new scaffold of small molecules with unique SARD and pan-antagonist activities even compared to our recently published AF-1 binding SARDs such as UT-155 (9) and UT-34 (10). Novel B-ring pyrazoles exhibited potent AR antagonist activities, including promising distribution, metabolism, and pharmacokinetic properties, and broad-spectrum AR antagonist properties, including potent in vivo antitumor activity. 26a was able to induce an 80% tumor growth inhibition of xenografts derived from the enzalutamide-resistant (Enz-R) VCaP cell line. These results represent an advancement toward the development of novel AR antagonists for the treatment of Enz-R prostate cancer.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Drug Resistance, Neoplasm/drug effects , Pyrazoles/chemistry , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/metabolism , Androgen Receptor Antagonists/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Half-Life , Humans , Male , Mice , Microsomes, Liver/metabolism , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Androgen/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
6.
PLoS One ; 15(3): e0230499, 2020.
Article in English | MEDLINE | ID: mdl-32187230

ABSTRACT

Glycosphingolipids (GSLs) hexosylceramides and lactosylceramides are elevated in lupus mice and human patients with nephritis. Whereas other renal diseases characterized by increased GSL levels are thought to be a result of upregulated GSL synthesis, our results suggest elevated hexosylceramides and lactosylceramides in lupus nephritis is a result of increased catabolism of ganglioside GM3 due to significantly increased neuraminidase (NEU) activity. Thus, we hypothesized GM3 would be decreased in lupus nephritis kidneys and blocking NEU activity would reduce GSLs and improve disease in lupus mice. Female MRL/lpr lupus mice were treated with water or the NEU inhibitor oseltamivir phosphate at the onset of proteinuria to block GSL catabolism. Age-matched (non-nephritic) female MRL/MpJ lupus mice served as controls. Renal GM3 levels were significantly higher in the nephritic MRL/lpr water-treated mice compared to non-nephritic MRL/MpJ mice, despite significantly increased renal NEU activity. Blocking GSL catabolism increased, rather than decreased, renal and urine GSL levels and disease was not significantly impacted. A pilot study treating MRL/lpr females with GlcCer synthase inhibitor Genz-667161 to block GSL synthesis resulted in a strong significant negative correlation between Genz-667161 dose and renal GSL hexosylceramide and GM3 levels. Splenomegaly was negatively correlated and serum IgG levels were marginally correlated with increasing Genz-667161 dose. These results suggest accumulation of renal GM3 may be due to dysregulation of one or more of the GSL ganglioside pathways and inhibiting GSL synthesis, but not catabolism, may be a therapeutic approach for treating lupus nephritis.


Subject(s)
Glycosphingolipids/metabolism , Lupus Nephritis/drug therapy , Lupus Nephritis/metabolism , Animals , Ceramides/metabolism , Female , G(M3) Ganglioside/metabolism , Kidney/drug effects , Kidney/metabolism , Lactosylceramides/metabolism , Mice , Mice, Inbred MRL lpr , Neuraminidase/metabolism , Oseltamivir/analogs & derivatives , Oseltamivir/therapeutic use , Phosphorous Acids/therapeutic use , Pilot Projects , Proteinuria/drug therapy , Proteinuria/metabolism
7.
iScience ; 21: 341-358, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31698248

ABSTRACT

Sustained treatment of estrogen receptor (ER)-positive breast cancer with ER-targeting drugs results in ER mutations and refractory unresponsive cancers. Androgen receptor (AR), which is expressed in 80%-95% of ER-positive breast cancers, could serve as an alternate therapeutic target. Although AR agonists were used in the past to treat breast cancer, their use is currently infrequent due to virilizing side effects. Discovery of tissue-selective AR modulators (SARMs) has renewed interest in using AR agonists to treat breast cancer. Using translational models, we show that AR agonist and SARM, but not antagonist, inhibit the proliferation and growth of ER-positive breast cancer cells, patient-derived tissues, and patient-derived xenografts (PDX). Ligand-activated AR inhibits wild-type and mutant ER activity by reprogramming the ER and FOXA1 cistrome and rendering tumor growth inhibition. These findings suggest that ligand-activated AR may function as a non-canonical inhibitor of ER and that AR agonists may offer a safe and effective treatment for ER-positive breast cancer.

8.
Clin Cancer Res ; 25(22): 6764-6780, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31481513

ABSTRACT

PURPOSE: Androgen receptor (AR)-targeting prostate cancer drugs, which are predominantly competitive ligand-binding domain (LBD)-binding antagonists, are inactivated by common resistance mechanisms. It is important to develop next-generation mechanistically distinct drugs to treat castration- and drug-resistant prostate cancers. EXPERIMENTAL DESIGN: Second-generation AR pan antagonist UT-34 was selected from a library of compounds and tested in competitive AR binding and transactivation assays. UT-34 was tested using biophysical methods for binding to the AR activation function-1 (AF-1) domain. Western blot, gene expression, and proliferation assays were performed in various AR-positive enzalutamide-sensitive and -resistant prostate cancer cell lines. Pharmacokinetic and xenograft studies were performed in immunocompromised rats and mice. RESULTS: UT-34 inhibits the wild-type and LBD-mutant ARs comparably and inhibits the in vitro proliferation and in vivo growth of enzalutamide-sensitive and -resistant prostate cancer xenografts. In preclinical models, UT-34 induced the regression of enzalutamide-resistant tumors at doses when the AR is degraded; but, at lower doses, when the AR is just antagonized, it inhibits, without shrinking, the tumors. This indicates that degradation might be a prerequisite for tumor regression. Mechanistically, UT-34 promotes a conformation that is distinct from the LBD-binding competitive antagonist enzalutamide and degrades the AR through the ubiquitin proteasome mechanism. UT-34 has a broad safety margin and exhibits no cross-reactivity with G-protein-coupled receptor kinase and nuclear receptor family members. CONCLUSIONS: Collectively, UT-34 exhibits the properties necessary for a next-generation prostate cancer drug.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/metabolism , Administration, Oral , Androgen Receptor Antagonists/administration & dosage , Androgen Receptor Antagonists/adverse effects , Androgen Receptor Antagonists/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Benzamides , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Gene Expression , Humans , Male , Mice , Mutation , Nitriles , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/adverse effects , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Rats , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Ubiquitin/metabolism , Xenograft Model Antitumor Assays
9.
Nanoscale Adv ; 1(9): 3392-3399, 2019 Sep 11.
Article in English | MEDLINE | ID: mdl-36133556

ABSTRACT

Tip-enhanced Raman spectroscopy (TERS) has been established as one the most efficient analytical techniques for probing vibrational states with nanoscale resolution. While TERS may be a source of unique information about chemical structure and interactions, it has a limited use for materials with rough or sticky surfaces. Development of the TERS approach utilizing a non-contact scanning probe microscopy mode can significantly extend the number of applications. Here we demonstrate a proof of the concept and feasibility of a non-contact TERS approach and test it on various materials. Our experiments show that non-contact TERS can provide 10 nm spatial resolution and a Raman signal enhancement factor of 105, making it very promising for chemical imaging of materials with high aspect ratio surface patterns and biomaterials.

10.
J Med Chem ; 62(2): 491-511, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30525603

ABSTRACT

In our effort to find small-molecule treatments of advanced prostate cancers (PCs), a novel series of indolyl and indolinyl propanamides (series II and III) were discovered as selective androgen receptor degraders (SARDs). Initial studies of androgen receptor (AR) antagonist (1) and agonist (2) propanamides yielded a tertiary aniline (3) with novel SARD activity but poor metabolic stability. Cyclization to II and III produced submicromolar AR antagonism and protein degradation selective to AR and AR splice variant (AR SV). II and III maintained potency against enzalutamide-resistant (Enz-R) mutant ARs and PC cells and were efficacious in Enz-R xenografts, suggesting their potential to treat advanced PCs. Design, synthesis, and biological activity of novel SARDs that could potentially be used for the treatment of a wide spectrum of PCs including castration-resistant, Enz-R, and/or AR SV-dependent advanced PCs that are often untreatable with known hormone therapies are discussed.


Subject(s)
Amides/chemistry , Drug Design , Drug Resistance, Neoplasm , Receptors, Androgen/metabolism , Amides/pharmacology , Amides/therapeutic use , Androgen Receptor Antagonists/chemistry , Androgens/chemistry , Animals , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Indoles/chemistry , Male , Mice , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Proteolysis , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
11.
Cancer Res ; 77(22): 6282-6298, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28978635

ABSTRACT

Androgen receptor (AR) mediates the growth of prostate cancer throughout its course of development, including in abnormal splice variants (AR-SV)-driven advanced stage castration-resistant disease. AR stabilization by androgens makes it distinct from other steroid receptors, which are typically ubiquitinated and degraded by proteasomes after ligand binding. Thus, targeting AR in advanced prostate cancer requires the development of agents that can sustainably degrade variant isoforms for effective therapy. Here we report the discovery and characterization of potent selective AR degraders (SARD) that markedly reduce the activity of wild-type and splice variant isoforms of AR at submicromolar doses. Three SARDs (UT-69, UT-155, and (R)-UT-155) bind the amino-terminal transcriptional activation domain AF-1, which has not been targeted for degradation previously, with two of these SARD (UT-69 and UT-155) also binding the carboxy-terminal ligand binding domain. Despite different mechanisms of action, all three SARDs degraded wild-type AR and inhibited AR function, exhibiting greater inhibitory potency than the approved AR antagonists. Collectively, our results introduce a new candidate class of next-generation therapeutics to manage advanced prostate cancer. Cancer Res; 77(22); 6282-98. ©2017 AACR.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/genetics , Alternative Splicing , Androgen Receptor Antagonists/chemistry , Anilides/chemistry , Anilides/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Gene Expression Profiling/methods , Humans , Indoles/chemistry , Indoles/pharmacology , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Molecular Structure , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays
12.
Hum Mol Genet ; 26(13): 2526-2540, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28453658

ABSTRACT

Duchenne muscular dystrophy (DMD) is a neuromuscular disease that predominantly affects boys as a result of mutation(s) in the dystrophin gene. DMD is characterized by musculoskeletal and cardiopulmonary complications, resulting in shorter life-span. Boys afflicted by DMD typically exhibit symptoms within 3-5 years of age and declining physical functions before attaining puberty. We hypothesized that rapidly deteriorating health of pre-pubertal boys with DMD could be due to diminished anabolic actions of androgens in muscle, and that intervention with an androgen receptor (AR) agonist will reverse musculoskeletal complications and extend survival. While castration of dystrophin and utrophin double mutant (mdx-dm) mice to mimic pre-pubertal nadir androgen condition resulted in premature death, maintenance of androgen levels extended the survival. Non-steroidal selective-AR modulator, GTx-026, which selectively builds muscle and bone was tested in X-linked muscular dystrophy mice (mdx). GTx-026 significantly increased body weight, lean mass and grip strength by 60-80% over vehicle-treated mdx mice. While vehicle-treated castrated mdx mice exhibited cardiopulmonary impairment and fibrosis of heart and lungs, GTx-026 returned cardiopulmonary function and intensity of fibrosis to healthy control levels. GTx-026 elicits its musculoskeletal effects through pathways that are distinct from dystrophin-regulated pathways, making AR agonists ideal candidates for combination approaches. While castration of mdx-dm mice resulted in weaker muscle and shorter survival, GTx-026 treatment increased the muscle mass, function and survival, indicating that androgens are important for extended survival. These preclinical results support the importance of androgens and the need for intervention with AR agonists to treat DMD-affected boys.


Subject(s)
Androgens/metabolism , Muscular Dystrophy, Duchenne/genetics , Androgens/genetics , Animals , Disease Models, Animal , Dystrophin/genetics , Fibrosis , Male , Mice , Mice, Inbred mdx , Muscle Weakness/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/metabolism , Receptors, Androgen/metabolism , Sexual Maturation , Utrophin/genetics
13.
Exp Biol Med (Maywood) ; 242(6): 606-616, 2017 03.
Article in English | MEDLINE | ID: mdl-28092182

ABSTRACT

Non-alcoholic steatohepatitis (NASH) affects 8-10 million people in the US and up to 75% of obese individuals. Despite this, there are no approved oral therapeutics to treat NASH and therefore the need for novel approaches exists. The estrogen receptor ß (ER-ß)-selective agonist, ß-LGND2, inhibits body weight and white adipose tissue, and increases metabolism, resulting in higher energy expenditure and thermogenesis. Due to favorable effects of ß-LGND2 on obesity, we hypothesized that ß-LGND2 will prevent NASH directly by reducing lipid accumulation in the liver or indirectly by favorably changing body composition. Male C57BL/6 mice fed with high fat diet (HFD) for 10 weeks or methionine choline-deficient diet for four weeks and treated with vehicle exhibited altered liver weights by twofold and increased serum transaminases by 2-6-folds. These changes were not observed in ß-LGND2-treated animals. Infiltration of inflammatory cells and collagen deposits, an indication of fibrosis, were observed in the liver of mice fed with HFD for 10 weeks, which were effectively blocked by ß-LGND2. Gene expression studies in the liver indicate that pregnane X receptor target genes were significantly increased by HFD, and the increase was inhibited by ß-LGND2. On the other hand, metabolomics indicate that bile acid metabolites were significantly increased by ß-LGND2. These studies demonstrate that an ER-ß agonist might provide therapeutic benefits in NASH by directly modulating the function of xenobiotic and bile acid receptors in the liver, which have important functions in the liver, and indirectly, as demonstrated before, by inhibiting adiposity. Impact statement Over 75-90% of those classified as clinically obese suffer from co-morbidities, the most common of which is non-alcoholic steatohepatitis (NASH). While there are currently no effective treatment approaches for NASH, data presented here provide preliminary evidence that an estrogen receptor ß-selective ligand could have the potential to reduce lipid accumulation and inflammation, and protect liver from NASH.


Subject(s)
Bile Acids and Salts/antagonists & inhibitors , Estrogen Receptor beta/agonists , Isoquinolines/therapeutic use , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Choline Deficiency/complications , Diet/adverse effects , Liver/pathology , Male , Metabolomics , Methionine/deficiency , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology
14.
J Cell Biochem ; 118(3): 640-646, 2017 03.
Article in English | MEDLINE | ID: mdl-27681158

ABSTRACT

Stress urinary incontinence (SUI), a prevalent condition, is represented by an involuntary leakage of urine that results, at least in part, from weakened or damaged pelvic floor muscles and is triggered by physical stress. Current treatment options are limited with no oral therapies available. The pelvic floor is rich in androgen receptor and molecules with anabolic activity including selective androgen receptor modulators (SARMs) may serve as therapeutic options for individuals with SUI. In this study, two SARMs (GTx-024 and GTx-027) were evaluated in a post-menopausal animal model in order to determine their effect on pelvic floor muscles. Female C57BL/6 mice were ovariectomized and their pelvic muscles allowed to regress. The animals were then treated with vehicle or doses of GTx-024 or GTx-027. Animal total body weight, lean body mass, and pelvic floor muscle weights were measured along with the expression of genes associated with muscle catabolism. Treatment with the SARMs resulted in a restoration of the pelvic muscles to the sham-operated weight. Coordinately, the induction of genes associated with muscle catabolism was inhibited. Although a trend was observed towards an increase in total lean body mass in the SARM-treated groups, no significant differences were detected. Treatment of an ovariectomized mouse model with SARMs resulted in an increase in pelvic floor muscles, which may translate to an improvement of symptoms associated with SUI and serves as the basis for evaluating their clinical use. J. Cell. Biochem. 118: 640-646, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Anilides/pharmacology , Muscle, Skeletal , Ovariectomy , Receptors, Androgen/metabolism , Urinary Incontinence, Stress , Animals , Female , HEK293 Cells , Humans , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Organ Size/drug effects , Pelvic Floor , Urinary Incontinence, Stress/drug therapy , Urinary Incontinence, Stress/metabolism , Urinary Incontinence, Stress/pathology , Urinary Incontinence, Stress/physiopathology
15.
FASEB J ; 31(1): 266-281, 2017 01.
Article in English | MEDLINE | ID: mdl-27733447

ABSTRACT

Most satiety-inducing obesity therapeutics, despite modest efficacy, have safety concerns that underscore the need for effective peripherally acting drugs. An attractive therapeutic approach for obesity is to optimize/maximize energy expenditure by increasing energy-utilizing thermogenic brown adipose tissue. We used in vivo and in vitro models to determine the role of estrogen receptor ß (ER-ß) and its ligands on adipose biology. RNA sequencing and metabolomics were used to determine the mechanism of action of ER-ß and its ligands. Estrogen receptor ß (ER-ß) and its selective ligand reprogrammed preadipocytes and precursor stem cells into brown adipose tissue and increased mitochondrial respiration. An ER-ß-selective ligand increased markers of tricarboxylic acid-dependent and -independent energy biogenesis and oxygen consumption in mice without a concomitant increase in physical activity or food consumption, all culminating in significantly reduced weight gain and adiposity. The antiobesity effects of ER-ß ligand were not observed in ER-ß-knockout mice. Serum metabolite profiles of adult lean and juvenile mice were comparable, while that of adult obese mice was distinct, indicating a possible impact of obesity on age-dependent metabolism. This phenotype was partially reversed by ER-ß-selective ligand. These data highlight a new role for ER-ß in adipose biology and its potential to be a safer alternative peripheral therapeutic target for obesity.-Ponnusamy, S., Tran, Q. T., Harvey, I., Smallwood, H. S., Thiyagarajan, T., Banerjee, S., Johnson, D. L., Dalton, J. T., Sullivan, R. D., Miller, D. D., Bridges, D., Narayanan, R. Pharmacologic activation of estrogen receptor ß increases mitochondrial function, energy expenditure, and brown adipose tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , Energy Metabolism/physiology , Estrogen Receptor beta/agonists , Estrogen Receptor beta/metabolism , Isoquinolines/pharmacology , Mitochondria/physiology , Adipose Tissue, White/physiology , Animals , Biomarkers , Diet, High-Fat , Estrogen Receptor beta/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Insulin Resistance , Male , Mice , Mice, Knockout , Obesity/blood , Obesity/metabolism
16.
Protein Expr Purif ; 123: 6-13, 2016 07.
Article in English | MEDLINE | ID: mdl-26965413

ABSTRACT

Telomerase biogenesis is a highly regulated process that solves the DNA end-replication problem. Recombinant expression has so far been accomplished only within a eukaryotic background. Towards structural and functional analyses, we developed bacterial expression of human telomerase. Positive activity by the telomerase repeat amplification protocol (TRAP) was identified in cell extracts of Escherichia coli expressing a sequence-optimized hTERT gene, the full-length hTR RNA with a self-splicing hepatitis delta virus ribozyme, and the human heat shock complex of Hsp90, Hsp70, p60/Hop, Hsp40, and p23. The Hsp90 inhibitor geldanamycin did not affect post-assembly TRAP activity. By various purification methods, TRAP activity was also obtained upon expression of only hTERT and hTR. hTERT was confirmed by tandem mass spectrometry in a ∼120 kDa SDS-PAGE fragment from a TRAP-positive purification fraction. TRAP activity was also supported by hTR constructs lacking the box H/ACA small nucleolar RNA domain. End-point TRAP indicated expression levels within 3-fold of that from HeLa carcinoma cells, which is several orders of magnitude below detection by the direct assay. These results represent the first report of TRAP activity from a bacterium and provide a facile system for the investigation of assembly factors and anti-cancer therapeutics independently of a eukaryotic setting.


Subject(s)
Escherichia coli/genetics , RNA/genetics , Telomerase/genetics , Amino Acid Sequence , Cloning, Molecular , DNA, Complementary/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Plasmids/genetics , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Telomerase/chemistry , Telomerase/isolation & purification , Telomerase/metabolism , Transformation, Genetic
17.
Oncol Rep ; 35(4): 2097-106, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26820391

ABSTRACT

Colon and pancreatic cancers contribute to 90,000 deaths each year in the USA. These cancers lack targeted therapeutics due to heterogeneity of the disease and multiple causative factors. One important factor that contributes to increased colon and pancreatic cancer risk is gastrin. Gastrin mediates its actions through two G-protein coupled receptors (GPCRs): cholecystokinin receptor A (CCK-A) and CCK-B/gastrin receptor. Previous studies have indicated that colon cancer predominantly expresses CCK-A and responds to CCK-A isoform antagonists. However, many CCK-A antagonists have failed in the clinic due to poor pharmacokinetic properties or lack of efficacy. In the present study, we synthesized a library of CCK-A isoform-selective antagonists and tested them in various colon and pancreatic cancer preclinical models. The lead CCK-A isoform, selective antagonist PNB-028, bound to CCK-A at 12 nM with a 60-fold selectivity towards CCK-A over CCK-B. Furthermore, it inhibited the proliferation of CCK-A-expressing colon and pancreatic cancer cells without affecting the proliferation of non-cancerous cells. PNB-028 was also extremely effective in inhibiting the growth of MAC-16 and LoVo colon cancer and MIA PaCa pancreatic cancer xenografts in immune-compromised mice. Genome­wide microarray and kinase-array studies indicate that PNB-028 inhibited oncogenic kinases and angiogenic factors to inhibit the growth of colon cancer xenografts. Safety pharmacology and toxicology studies have indicated that PNB-028 is extremely safe and has a wide safety margin. These studies suggest that targeting CCK-A selectively renders promise to treat colon and pancreatic cancers and that PNB-028 could become the next-generation treatment option.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Antineoplastic Agents/administration & dosage , Colonic Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/chemical synthesis , Receptor, Cholecystokinin A/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , COS Cells , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorocebus aethiops , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Drug Evaluation, Preclinical , HCT116 Cells , HT29 Cells , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Peptide Library , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Receptor, Cholecystokinin A/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
18.
J Immunol ; 195(12): 5551-60, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26538397

ABSTRACT

The ETS factor Friend leukemia virus integration 1 (FLI1) is a key modulator of lupus disease expression. Overexpressing FLI1 in healthy mice results in the development of an autoimmune kidney disease similar to that observed in lupus. Lowering the global levels of FLI1 in two lupus strains (Fli1(+/-)) significantly improved kidney disease and prolonged survival. T cells from MRL/lpr Fli1(+/-) lupus mice have reduced activation and IL-4 production, neuraminidase 1 expression, and the levels of the glycosphingolipid lactosylceramide. In this study, we demonstrate that MRL/lpr Fli1(+/-) mice have significantly decreased renal neuraminidase 1 and lactosylceramide levels. This corresponds with a significant decrease in the number of total CD3(+) cells, as well as CD4(+) and CD44(+)CD62L(-) T cell subsets in the kidney of MRL/lpr Fli1(+/-) mice compared with the Fli1(+/+) nephritic mice. We further demonstrate that the percentage of CXCR3(+) T cells and Cxcr3 message levels in T cells are significantly decreased and correspond with a decrease in renal CXCR3(+) cells and in Cxcl9 and Cxcl10 expression in the MRL/lpr Fli1(+/-) compared with the Fli1(+/+) nephritic mice. Our results suggest that reducing the levels of FLI1 in MRL/lpr mice may be protective against development of nephritis in part through downregulation of CXCR3, reducing renal T cell infiltration and glycosphingolipid levels.


Subject(s)
Glycosphingolipids/metabolism , Kidney/physiology , Nephritis/drug therapy , Proto-Oncogene Protein c-fli-1/metabolism , Receptors, CXCR3/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , Cell Movement/drug effects , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Gene Expression Regulation , Humans , Kidney/drug effects , Lactosylceramides/metabolism , Mice , Mice, Inbred MRL lpr , Mice, Knockout , Nephritis/immunology , Neuraminidase/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Receptors, CXCR3/genetics
19.
J Am Soc Nephrol ; 26(6): 1402-13, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25270066

ABSTRACT

Nearly one half of patients with lupus develop glomerulonephritis (GN), which often leads to renal failure. Although nephritis is diagnosed by the presence of proteinuria, the pathology of nephritis can fall into one of five classes defined by different forms of tissue injury, and the mechanisms involved in pathogenesis are not completely understood. Glycosphingolipids are abundant in the kidney, have roles in many cellular functions, and were shown to be involved in other renal diseases. Here, we show dysfunctional glycosphingolipid metabolism in patients with lupus nephritis and MRL/lpr lupus mice. Specifically, we found that glucosylceramide (GlcCer) and lactosylceramide (LacCer) levels are significantly higher in the kidneys of nephritic MRL/lpr lupus mice than the kidneys of non-nephritic lupus mice or healthy controls. This elevation may be, in part, caused by altered transcriptional regulation and/or activity of LacCer synthase (GalT5) and neuraminidase 1, enzymes that mediate glycosphingolipid metabolism. We show increased neuraminidase 1 activity early during the progression of nephritis (before significant elevation of GlcCer and LacCer in the kidney). Elevated levels of urinary LacCer were detected before proteinuria in lupus mice. Notably, LacCer levels were higher in the urine and kidneys of patients with lupus and nephritis than patients with lupus without nephritis or healthy controls. Together, these results show early and significant dysfunction of the glycosphingolipid metabolic pathway in the kidneys of lupus mice and patients with lupus nephritis and suggest that molecules in this pathway may serve as early markers in lupus nephritis.


Subject(s)
Glycosphingolipids/metabolism , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Neuraminidase/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Analysis of Variance , Animals , Biomarkers/analysis , Biopsy, Needle , Disease Models, Animal , Disease Progression , Follow-Up Studies , Humans , Immunoblotting , Immunohistochemistry , Kidney Function Tests , Lupus Nephritis/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Neuraminidase/genetics , Sensitivity and Specificity , Severity of Illness Index , Sterol Regulatory Element Binding Protein 1/genetics , Urinalysis
20.
PLoS One ; 8(9): e75175, 2013.
Article in English | MEDLINE | ID: mdl-24040398

ABSTRACT

Systemic Lupus erythematosus (SLE) is an autoimmune disease caused, in part, by abnormalities in cells of the immune system including B and T cells. Genetically reducing globally the expression of the ETS transcription factor FLI1 by 50% in two lupus mouse models significantly improves disease measures and survival through an unknown mechanism. In this study we analyze the effects of reducing FLI1 in the MRL/lpr lupus prone model on T cell function. We demonstrate that adoptive transfer of MRL/lpr Fli1(+/+) or Fli1(+/-) T cells and B cells into Rag1-deficient mice results in significantly decreased serum immunoglobulin levels in animals receiving Fli1(+/-) lupus T cells compared to animals receiving Fli1(+/+) lupus T cells regardless of the genotype of co-transferred lupus B cells. Ex vivo analyses of MRL/lpr T cells demonstrated that Fli1(+/-) T cells produce significantly less IL-4 during early and late disease and exhibited significantly decreased TCR-specific activation during early disease compared to Fli1(+/+) T cells. Moreover, the Fli1(+/-) T cells expressed significantly less neuraminidase 1 (Neu1) message and decreased NEU activity during early disease and significantly decreased levels of glycosphingolipids during late disease compared to Fli1(+/+) T cells. FLI1 dose-dependently activated the Neu1 promoter in mouse and human T cell lines. Together, our results suggest reducing FLI1 in lupus decreases the pathogenicity of T cells by decreasing TCR-specific activation and IL-4 production in part through the modulation of glycosphingolipid metabolism. Reducing the expression of FLI1 or targeting the glycosphingolipid metabolic pathway in lupus may serve as a therapeutic approach to treating lupus.


Subject(s)
Glycosphingolipids/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adoptive Transfer , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Models, Animal , Disease Progression , Female , Homeodomain Proteins/metabolism , Humans , Interleukin-4/biosynthesis , Jurkat Cells , Lymphocyte Activation , Mice , Mice, Inbred MRL lpr , Proto-Oncogene Protein c-fli-1/deficiency , Receptors, Antigen, T-Cell/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...