Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Pharmacol Rev ; 75(6): 1233-1318, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37586884

ABSTRACT

The NR superfamily comprises 48 transcription factors in humans that control a plethora of gene network programs involved in a wide range of physiologic processes. This review will summarize and discuss recent progress in NR biology and drug development derived from integrating various approaches, including biophysical techniques, structural studies, and translational investigation. We also highlight how defective NR signaling results in various diseases and disorders and how NRs can be targeted for therapeutic intervention via modulation via binding to synthetic lipophilic ligands. Furthermore, we also review recent studies that improved our understanding of NR structure and signaling. SIGNIFICANCE STATEMENT: Nuclear receptors (NRs) are ligand-regulated transcription factors that are critical regulators of myriad physiological processes. NRs serve as receptors for an array of drugs, and in this review, we provide an update on recent research into the roles of these drug targets.


Subject(s)
Pharmacology, Clinical , Humans , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Carrier Proteins , Ligands
2.
Mo Med ; 118(5): 466-472, 2021.
Article in English | MEDLINE | ID: mdl-34658442

ABSTRACT

The leading cause blindness is the loss of retinal ganglion cells which connect the retina to the brain. Degenerative retinal diseases include retinal dystrophy, macular degeneration and diabetic retinopathy, which are currently incurable as the mammalian retina has no intrinsic regenerative capacity. By utilizing insight gained from retinal regeneration in simpler species we define an approach that may unlock regenerative programs in the mammalian retina that potentially facilitate the clinical restoration of retinal function.


Subject(s)
Retinal Degeneration , Humans , Retinal Degeneration/therapy
3.
PLoS One ; 16(3): e0249316, 2021.
Article in English | MEDLINE | ID: mdl-33770118

ABSTRACT

Alcoholic liver disease (ALD) is responsible for an average of 50.4% and 44.2%of liver disease deaths among males and females respectively. Driven by alcohol misuse, ALD is often reversible by cessation of consumption. However, abstinence programs can have limited success at curtailing abuse, and the loss of life. ALD, therefore, remains a significant clinical challenge. There is a need for effective treatments that prevent or reverse alcohol-induced liver damage to complement or supplant behavioral interventions. Metabolic syndrome, which is disproportionally prevalent in ALD patients, accelerates the progression of ALD and increases liver disease mortality. Current rodent models of ALD unfortunately do not account for the contribution of the western diet to ALD pathology. To address this, we have developed a rodent model of ALD that integrates the impact of the western diet and alcohol; the WASH-diet model. We show here that the WASH diet, either chronically or in small time-restricted bouts, accelerated ALD pathology with severe steatohepatitis, elevated inflammation and increased fibrosis compared to mice receiving chronic alcohol alone. We also validated our WASH-diet model as an in vivo system for testing the efficacy of experimental ALD treatments. The efficacy of the inverse-agonist SR9238, previously shown to inhibit both non-alcohol and alcohol-induced steatohepatitis progression, was conserved in our WASH-diet model. These findings suggested that the WASH-diet may be useful for in vivo pre-clinical assessment of novel therapies.


Subject(s)
Liver Cirrhosis/complications , Liver Diseases, Alcoholic/complications , Animals , Diet, Western , Female , Male , Mice , Models, Biological , Time Factors
4.
Cell Rep ; 34(8): 108768, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33626346

ABSTRACT

Mucoepidermoid carcinoma (MEC) is a life-threatening salivary gland cancer that is driven primarily by a transcriptional coactivator fusion composed of cyclic AMP-regulated transcriptional coactivator 1 (CRTC1) and mastermind-like 2 (MAML2). The mechanisms by which the chimeric CRTC1/MAML2 (C1/M2) oncoprotein rewires gene expression programs that promote tumorigenesis remain poorly understood. Here, we show that C1/M2 induces transcriptional activation of the non-canonical peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) splice variant PGC-1α4, which regulates peroxisome proliferator-activated receptor gamma (PPARγ)-mediated insulin-like growth factor 1 (IGF-1) expression. This mitogenic transcriptional circuitry is consistent across cell lines and primary tumors. C1/M2-positive tumors exhibit IGF-1 pathway activation, and small-molecule drug screens reveal that tumor cells harboring the fusion gene are selectively sensitive to IGF-1 receptor (IGF-1R) inhibition. Furthermore, this dependence on autocrine regulation of IGF-1 transcription renders MEC cells susceptible to PPARγ inhibition with inverse agonists. These results yield insights into the aberrant coregulatory functions of C1/M2 and identify a specific vulnerability that can be exploited for precision therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Mucoepidermoid/drug therapy , Insulin-Like Growth Factor I/metabolism , PPAR gamma/antagonists & inhibitors , Salivary Gland Neoplasms/drug therapy , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Autocrine Communication , Carcinoma, Mucoepidermoid/genetics , Carcinoma, Mucoepidermoid/metabolism , Carcinoma, Mucoepidermoid/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Gene Fusion , Humans , Insulin-Like Growth Factor I/genetics , Male , Mice, Nude , Middle Aged , Molecular Targeted Therapy , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Isoforms , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/metabolism , Salivary Gland Neoplasms/pathology , Signal Transduction , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
PLoS One ; 15(5): e0227720, 2020.
Article in English | MEDLINE | ID: mdl-32407314

ABSTRACT

Numerous mutational studies have demonstrated that circadian clock proteins regulate behavior and metabolism. Nr1d1(Rev-erbα) is a key regulator of circadian gene expression and a pleiotropic regulator of skeletal muscle homeostasis and lipid metabolism. Loss of Rev-erbα expression induces muscular atrophy, high adiposity, and metabolic syndrome in mice. Here we show that, unlike knockout mice, Nr1d1 heterozygous mice are not susceptible to muscular atrophy and in fact paradoxically possess larger myofiber diameters and improved neuromuscular function, compared to wildtype mice. Heterozygous mice lacked dyslipidemia, a characteristic of Nr1d1 knockout mice and displayed increased whole-body fatty-acid oxidation during periods of inactivity (light cycle). Heterozygous mice also exhibited higher rates of glucose uptake when fasted, and had elevated basal rates of gluconeogenesis compared to wildtype and knockout littermates. Rev-erbα ablation suppressed glycolysis and fatty acid-oxidation in white-adipose tissue (WAT), whereas partial Rev-erbα loss, curiously stimulated these processes. Our investigations revealed that Rev-erbα dose-dependently regulates glucose metabolism and fatty acid oxidation in WAT and muscle.


Subject(s)
Dyslipidemias/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Adipose Tissue, White/metabolism , Adiposity/genetics , Animals , Behavior, Animal/physiology , Circadian Clocks/genetics , Dyslipidemias/metabolism , Dyslipidemias/pathology , Fatty Acids/metabolism , Gluconeogenesis/genetics , Glucose/metabolism , Heterozygote , Humans , Lipid Metabolism/genetics , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Mice , Mice, Knockout , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myofibrils/genetics , Myofibrils/metabolism , Myofibrils/pathology , Photoperiod
6.
Sci Rep ; 9(1): 19530, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31863071

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive subtype that is untreatable with hormonal or HER2-targeted therapies and is also typically unresponsive to checkpoint-blockade immunotherapy. Within the tumor microenvironment dysregulated immune cell metabolism has emerged as a key mechanism of tumor immune-evasion. We have discovered that the Liver-X-Receptors (LXRα and LXRß), nuclear receptors known to regulate lipid metabolism and tumor-immune interaction, are highly activated in TNBC tumor associated myeloid cells. We therefore theorized that inhibiting LXR would induce immune-mediated TNBC-tumor clearance. Here we show that pharmacological inhibition of LXR activity induces tumor destruction primarily through stimulation of CD8+ T-cell cytotoxic activity and mitochondrial metabolism. Our results imply that LXR inverse agonists may be a promising new class of TNBC immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/metabolism , Triple Negative Breast Neoplasms/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/physiology , Xenograft Model Antitumor Assays
7.
ACS Pharmacol Transl Sci ; 1(1): 50-60, 2018 Sep 14.
Article in English | MEDLINE | ID: mdl-31696159

ABSTRACT

Alcohol abuse is a major cause of liver disease and mortality worldwide and is a significant public health issue. Patients with alcoholic liver disease (ALD) have severe hepatic lipid accumulation, inflammation, and fibrosis. Therapies for ALD are very limited and even abstinence from alcohol consumption does not necessarily protect patients from progression of the disease. We sought to evaluate the efficacy of a liver X receptor (LXR) inverse agonist, SR9238, in an animal model of ALD. SR9238 suppresses hepatic lipogenesis, a pathological hallmark of ALD, and we hypothesized that targeting suppression of hepatic metabolic pathways that are activated in ALD may be an effective treatment for the disease. A chronic ethanol diet with or without a final ethanol binge treatment was used to induce ALD in mice. Mice were administered the liver specific LXR inverse agonist SR9238 for 4 weeks after the mice had been maintained on the ethanol diet for 14 days. Mice developed all the hallmarks of advanced ALD demonstrating significant pathophysiology and hepatotoxicity. SR9238 significantly attenuated liver injury and hepatic steatosis and fibrosis was nearly eliminated in SR9238 treated mice. SR9238 treatment reversed the damage associated with chronic ethanol use returning the liver to near normal morphology. These results indicate that inhibiting LXR activity using the inverse agonist has a hepatoprotective effect in rodent models of ALD; thus, this pharmacological approach may be efficacious for treatment of ALD in humans.

8.
Sci Rep ; 7(1): 17142, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215066

ABSTRACT

Duchenne muscular dystrophy (DMD) is a debilitating X-linked disorder that is fatal. DMD patients lack the expression of the structural protein dystrophin caused by mutations within the DMD gene. The absence of functional dystrophin protein results in excessive damage from normal muscle use due to the compromised structural integrity of the dystrophin associated glycoprotein complex. As a result, DMD patients exhibit ongoing cycles of muscle destruction and regeneration that promote inflammation, fibrosis, mitochondrial dysfunction, satellite cell (SC) exhaustion and loss of skeletal and cardiac muscle function. The nuclear receptor REV-ERB suppresses myoblast differentiation and recently we have demonstrated that the REV-ERB antagonist, SR8278, stimulates muscle regeneration after acute injury. Therefore, we decided to explore whether the REV-ERB antagonist SR8278 could slow the progression of muscular dystrophy. In mdx mice SR8278 increased lean mass and muscle function, and decreased muscle fibrosis and muscle protein degradation. Interestingly, we also found that SR8278 increased the SC pool through stimulation of Notch and Wnt signaling. These results suggest that REV-ERB is a potent target for the treatment of DMD.


Subject(s)
Cell Differentiation/drug effects , Fibrosis/prevention & control , Isoquinolines/pharmacology , Muscle, Skeletal/cytology , Muscular Dystrophy, Animal/complications , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , Regeneration , Thiophenes/pharmacology , Animals , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Receptors, Notch/metabolism , Signal Transduction , Wnt Proteins/metabolism
9.
Mol Metab ; 6(7): 703-714, 2017 07.
Article in English | MEDLINE | ID: mdl-28702326

ABSTRACT

OBJECTIVE: The loss of skeletal muscle mass and strength are a central feature of traumatic injury and degenerative myopathies. Unfortunately, pharmacological interventions typically fail to stem the long-term decline in quality of life. Reduced Rev-Erb-mediated gene suppression in cultured C2C12 myoblasts has been shown to stimulate myoblast differentiation. Yet the mechanisms that allow Rev-Erb to pleiotropically inhibit muscle differentiation are not well understood. In this study, we sought to elucidate the role of Rev-Erb in the regulation of muscle differentiation and regeneration in vivo. METHODS: Using Rev-Erbα/ß shRNAs, pharmacological ligands, and Rev-Erbα null and heterozygous mice, we probed the mechanism of Rev-Erbα/ß regulation of muscle differentiation and muscle regeneration. RESULTS: ChIP seq analysis of Rev-Erb in differentiating myoblasts showed that Rev-Erbα did not transcriptionally regulate muscle differentiation through cognate Rev-Erb/ROR-response elements but through possible interaction with the cell fate regulator NF-Y at CCAAT-motifs. Muscle differentiation is stimulated by Rev-Erb release from CCAAT-motifs at promoter and enhancer elements of a number of myogenesis proteins. Partial loss of Rev-Erb expression in mice heterozygous for Rev-Erbα accelerated muscle repair in vivo whereas Rev-Erb knockout mice showed deficiencies in regenerative repair compared to wild type mice. These phenotypic differences between heterozygous and knockout mice were not apparently dependent on MRF induction in response to injury. Similarly, pharmacological disruption of Rev-Erb suppressive activity in injured muscle accelerated regenerative repair in response to acute injury. CONCLUSIONS: Disrupting Rev-Erb activity in injured muscle accelerates regenerative muscle repair/differentiation through transcriptional de-repression of myogenic programs. Rev-Erb, therefore, may be a potent therapeutic target for a myriad of muscular disorders.


Subject(s)
CCAAT-Binding Factor/metabolism , Muscular Atrophy/metabolism , Myoblasts, Skeletal/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Regeneration , Adult , Animals , CCAAT-Binding Factor/genetics , Cell Differentiation , Cells, Cultured , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/physiology , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
10.
Phys Biol ; 14(4): 045002, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28586319

ABSTRACT

Muscle is primarily known for its mechanical roles in locomotion, maintenance of posture, and regulation of cardiac and respiratory function. There are numerous medical conditions that adversely affect muscle, myopathies that disrupt muscle development, regeneration and protein turnover to detrimental effect. Skeletal muscle is also a vital secretory organ that regulates thermogenesis, inflammatory signaling and directs context specific global metabolic changes in energy substrate preference on a daily basis. Myopathies differ in the causative factors that drive them but share common features including severe reduction in quality of life and significantly increased mortality all due irrefutably to the loss of muscle mass. Thus far clinically viable approaches for preserving muscle proteins and stimulating new muscle growth without unwanted side effects or limited efficacy has been elusive. Over the last few decades, evidence has emerged through in vitro and in vivo studies that suggest the nuclear receptors REV-ERB and ROR might modulate pathways involved in myogenesis and mitochondrial biogenesis. Hinting that REV-ERB and ROR might be targeted to treat myopathies. However there is still a need for substantial investigation into the roles of these nuclear receptors in in vivo rodent models of degenerative muscle diseases and acute injury. Although exciting, REV-ERB and ROR have somewhat confounding roles in muscle physiology and therefore more studies utilizing in vivo models of skeletal muscle myopathies are needed. In this review we highlight the molecular forces driving some of the major degenerative muscular diseases and showcase two promising molecular targets that may have the potential to treat myopathies: ROR and REV-ERB.


Subject(s)
Molecular Targeted Therapy/methods , Muscle, Skeletal/physiology , Muscular Diseases/metabolism , Muscular Diseases/therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Diseases/physiopathology , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Repressor Proteins/metabolism , Signal Transduction
11.
Cancer Res ; 75(23): 5023-33, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26424696

ABSTRACT

Fluorescent proteins are widely used to study molecular and cellular events, yet this traditionally relies on delivery of excitation light, which can trigger autofluorescence, photoxicity, and photobleaching, impairing their use in vivo. Accordingly, chemiluminescent light sources such as those generated by luciferases have emerged, as they do not require excitation light. However, current luciferase reporters lack the brightness needed to visualize events in deep tissues. We report the creation of chimeric eGFP-NanoLuc (GpNLuc) and LSSmOrange-NanoLuc (OgNLuc) fusion reporter proteins coined LumiFluors, which combine the benefits of eGFP or LSSmOrange fluorescent proteins with the bright, glow-type bioluminescent light generated by an enhanced small luciferase subunit (NanoLuc) of the deep-sea shrimp Oplophorus gracilirostris. The intramolecular bioluminescence resonance energy transfer that occurs between NanoLuc and the fused fluorophore generates the brightest bioluminescent signal known to date, including improved intensity, sensitivity, and durable spectral properties, thereby dramatically reducing image acquisition times and permitting highly sensitive in vivo imaging. Notably, the self-illuminating and bifunctional nature of these LumiFluor reporters enables greatly improved spatiotemporal monitoring of very small numbers of tumor cells via in vivo optical imaging and also allows the isolation and analyses of single cells by flow cytometry. Thus, LumiFluor reporters are inexpensive, robust, noninvasive tools that allow for markedly improved in vivo optical imaging of tumorigenic processes.


Subject(s)
Carcinogenesis/chemistry , Flow Cytometry/methods , Green Fluorescent Proteins/chemistry , Luciferases/chemistry , Luminescent Agents/chemistry , Optical Imaging/methods , Recombinant Fusion Proteins/chemistry , Animals , Burkitt Lymphoma/chemistry , Burkitt Lymphoma/pathology , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/pathology , Decapoda/enzymology , Green Fluorescent Proteins/genetics , HEK293 Cells , Heterografts , Humans , Luciferases/genetics , Lung Neoplasms/chemistry , Lung Neoplasms/pathology , Mice, Inbred NOD , Mice, SCID , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/genetics
12.
Cancer Cell ; 28(1): 42-56, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26120082

ABSTRACT

Malignant cells exhibit aerobic glycolysis (the Warburg effect) and become dependent on de novo lipogenesis, which sustains rapid proliferation and resistance to cellular stress. The nuclear receptor liver-X-receptor (LXR) directly regulates expression of key glycolytic and lipogenic genes. To disrupt these oncogenic metabolism pathways, we designed an LXR inverse agonist SR9243 that induces LXR-corepressor interaction. In cancer cells, SR9243 significantly inhibited the Warburg effect and lipogenesis by reducing glycolytic and lipogenic gene expression. SR9243 induced apoptosis in tumors without inducing weight loss, hepatotoxicity, or inflammation. Our results suggest that LXR inverse agonists may be an effective cancer treatment approach.


Subject(s)
Antineoplastic Agents/administration & dosage , Lipogenesis/drug effects , Neoplasms/drug therapy , Orphan Nuclear Receptors/agonists , Small Molecule Libraries/administration & dosage , Sulfonamides/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Glycolysis/drug effects , HT29 Cells , Hep G2 Cells , Humans , Liver X Receptors , Mice , Molecular Targeted Therapy , Neoplasms/pathology , Organ Specificity , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Weight Loss/drug effects , Xenograft Model Antitumor Assays
13.
Mol Metab ; 4(4): 353-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25830098

ABSTRACT

OBJECTIVE: Non-alcoholic steatohepatitis (NASH) is characterized by hepatic steatosis, inflammation and fibrosis. There are currently no targeted therapies for NASH. We developed a liver-specific LXR inverse agonist, SR9238, which effectively reduces hepatic lipogenesis in models of obesity and hepatic steatosis. We hypothesized that suppression of lipogenesis, which is pathologically elevated in NASH may suppress progression of hepatic steatosis to NASH. METHODS: NASH was induced in B6 V-lep (ob)/J (ob/ob) mice using a custom complete rodent diet (HTF) containing high amounts of trans-fat, fructose, and cholesterol. Once NASH was induced, mice were treated with SR9238 for one month by i.p. injection. Plasma lipid levels and liver health were analyzed by clinical chemistry. QPCR, western blot, and immunohistochemistry were used to assess disease severity. RESULTS: Ob/ob mice are obese and diabetic thus they are commonly used as models for the study of metabolic diseases. These mice quickly developed the NASH phenotype when provided the HTF diet. The mice develop hepatic steatosis, severe hepatic inflammation and fibrosis on the HTF diet. Treatment with SR9238 significantly reduced the severity of hepatic steatosis and most importantly reduced hepatic inflammation and ameliorated hepatic fibrosis. CONCLUSIONS: Here, we demonstrate that an LXR inverse agonist, SR9238, is effective in reduction of hepatic steatosis, inflammation and fibrosis in an animal model of NASH. These results have important implications for the development of therapeutics for treatment NASH in humans.

14.
Cancer Res ; 74(17): 4811-21, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24994715

ABSTRACT

The Hedgehog (HH) signaling pathway represents an important class of emerging developmental signaling pathways that play critical roles in the genesis of a large number of human cancers. The pharmaceutical industry is currently focused on developing small molecules targeting Smoothened (Smo), a key signaling effector of the HH pathway that regulates the levels and activity of the Gli family of transcription factors. Although one of these compounds, vismodegib, is now FDA-approved for patients with advanced basal cell carcinoma, acquired mutations in Smo can result in rapid relapse. Furthermore, many cancers also exhibit a Smo-independent activation of Gli proteins, an observation that may underlie the limited efficacy of Smo inhibitors in clinical trials against other types of cancer. Thus, there remains a critical need for HH inhibitors with different mechanisms of action, particularly those that act downstream of Smo. Recently, we identified the FDA-approved anti-pinworm compound pyrvinium as a novel, potent (IC50, 10 nmol/L) casein kinase-1α (CK1α) agonist. We show here that pyrvinium is a potent inhibitor of HH signaling, which acts by reducing the stability of the Gli family of transcription factors. Consistent with CK1α agonists acting on these most distal components of the HH signaling pathway, pyrvinium is able to inhibit the activity of a clinically relevant, vismodegib -resistant Smo mutant, as well as the Gli activity resulting from loss of the negative regulator suppressor of fused. We go on to demonstrate the utility of this small molecule in vivo, against the HH-dependent cancer medulloblastoma, attenuating its growth and reducing the expression of HH biomarkers.


Subject(s)
Hedgehog Proteins/metabolism , Pyrvinium Compounds/pharmacology , Signal Transduction/drug effects , Animals , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/metabolism , Casein Kinase Ialpha/metabolism , Cell Line , HEK293 Cells , Humans , Medulloblastoma/drug therapy , Medulloblastoma/metabolism , Mice , Mice, Nude , NIH 3T3 Cells , Oncogene Proteins , Receptors, G-Protein-Coupled/metabolism , Trans-Activators , Transcription Factors/metabolism , Zinc Finger Protein GLI1
15.
PLoS One ; 9(7): e101969, 2014.
Article in English | MEDLINE | ID: mdl-25003333

ABSTRACT

Mutations in the WNT-pathway regulator ADENOMATOUS POLYPOSIS COLI (APC) promote aberrant activation of the WNT pathway that is responsible for APC-associated diseases such as Familial Adenomatous Polyposis (FAP) and 85% of spontaneous colorectal cancers (CRC). FAP is characterized by multiple intestinal adenomas, which inexorably result in CRC. Surprisingly, given their common occurrence, there are few effective chemotherapeutic drugs for FAP. Here we show that the FDA-approved, anti-helminthic drug Pyrvinium attenuates the growth of WNT-dependent CRC cells and does so via activation of CK1α. Furthermore, we show that Pyrvinium can function as an in vivo inhibitor of WNT-signaling and polyposis in a mouse model of FAP: APCmin mice. Oral administration of Pyrvinium, a CK1α agonist, attenuated the levels of WNT-driven biomarkers and inhibited adenoma formation in APCmin mice. Considering its well-documented safe use for treating enterobiasis in humans, our findings suggest that Pyrvinium could be repurposed for the clinical treatment of APC-associated polyposes.


Subject(s)
Adenomatous Polyposis Coli/drug therapy , Antineoplastic Agents/pharmacology , Pyrvinium Compounds/pharmacology , Animals , Cell Survival/drug effects , Drug Approval , Drug Repositioning , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Mice, Inbred C57BL , Mice, Transgenic , Wnt Signaling Pathway/drug effects
16.
Mol Pharmacol ; 79(3): 508-19, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21127131

ABSTRACT

We have characterized previously a class of aryl hydrocarbon receptor (AHR) ligand termed selective AHR modulators (SAhRMs). SAhRMs exhibit anti-inflammatory properties, including suppression of cytokine-mediated acute phase genes (e.g., Saa1), through dissociation of non-dioxin-response element (DRE) AHR activity from DRE-dependent xenobiotic gene expression. The partial AHR agonist α-naphthoflavone (αNF) mediates the suppressive, non-DRE dependent effects on SAA1 expression and partial DRE-mediated CYP1A1 induction. These observations suggest that αNF may be structurally modified to a derivative exhibiting only SAhRM activity. A screen of αNF derivatives identifies 3',4'-dimethoxy-αNF (DiMNF) as a candidate SAhRM. Competitive ligand binding validates DiMNF as an AHR ligand, and DRE-dependent reporter assays with quantitative mRNA analysis of AHR target genes reveal minimal agonist activity associated with AHR binding. Consistent with loss of agonist activity, DiMNF fails to promote AHR binding to DRE probes as determined through electromobility shift assay. Importantly, mRNA analysis indicates that DiMNF retains the suppressive capacity of αNF regarding cytokine-mediated SAA1 expression in Huh7 cells. Interestingly, predictive docking modeling suggests that DiMNF adopts a unique orientation within the AHR ligand binding pocket relative to αNF and may facilitate the rational design of additional SAhRMs. Microarray studies with a non-DRE binding but otherwise functional AHR mutant identified complement factor C3 as a potential SAhRM target. We confirmed this observation in Huh7 cells using 10 µM DiMNF, which significantly repressed C3 mRNA and protein. These data expand the classes of AHR ligands exerting DRE-independent anti-inflammatory SAhRM activity, suggesting SAhRMs may have application in the amelioration of inflammatory disorders.


Subject(s)
Benzoflavones/pharmacology , Complement C3/biosynthesis , Cytokines/physiology , Receptors, Aryl Hydrocarbon/drug effects , Acute-Phase Reaction/metabolism , Cell Line , Complement C3/genetics , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Gene Expression/drug effects , Humans , Inflammation/drug therapy , Inflammation/metabolism , Ligands , Photoaffinity Labels/metabolism , Protein Binding/drug effects , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serum Amyloid A Protein/metabolism
17.
Science ; 329(5997): 1345-8, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20688981

ABSTRACT

Although practiced clinically for more than 40 years, the use of hematopoietic stem cell (HSC) transplants remains limited by the ability to expand these cells ex vivo. An unbiased screen with primary human HSCs identified a purine derivative, StemRegenin 1 (SR1), that promotes the ex vivo expansion of CD34+ cells. Culture of HSCs with SR1 led to a 50-fold increase in cells expressing CD34 and a 17-fold increase in cells that retain the ability to engraft immunodeficient mice. Mechanistic studies show that SR1 acts by antagonizing the aryl hydrocarbon receptor (AHR). The identification of SR1 and AHR modulation as a means to induce ex vivo HSC expansion should facilitate the clinical use of HSC therapy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Purines/metabolism , Purines/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , AC133 Antigen , Animals , Antigens, CD/analysis , Antigens, CD34/analysis , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cell Count , Cell Lineage , Cell Proliferation , Cells, Cultured , Cytochrome P-450 CYP1B1 , Cytokines/pharmacology , Glycoproteins/analysis , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/physiology , Peptides/analysis , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Small Molecule Libraries , Species Specificity
18.
Toxicol Sci ; 115(1): 89-97, 2010 May.
Article in English | MEDLINE | ID: mdl-20106948

ABSTRACT

Inflammatory signaling plays a key role in tumor progression, and the pleiotropic cytokine interleukin-6 (IL-6) is an important mediator of protumorigenic properties. Activation of the aryl hydrocarbon receptor (AHR) with exogenous ligands coupled with inflammatory signals can lead to synergistic induction of IL6 expression in tumor cells. Whether there are endogenous AHR ligands that can mediate IL6 production remains to be established. The indoleamine-2,3-dioxygenase pathway is a tryptophan oxidation pathway that is involved in controlling immune tolerance, which also aids in tumor escape. We screened the metabolites of this pathway for their ability to activate the AHR; results revealed that kynurenic acid (KA) is an efficient agonist for the human AHR. Structure-activity studies further indicate that the carboxylic acid group is required for significant agonist activity. KA is capable of inducing CYP1A1 messenger RNA levels in HepG2 cells and inducing CYP1A-mediated metabolism in primary human hepatocytes. In a human dioxin response element-driven stable reporter cell line, the EC(25) was observed to be 104nM, while in a mouse stable reporter cell line, the EC(25) was 10muM. AHR ligand competition binding assays revealed that KA is a ligand for the AHR. Treatment of MCF-7 cells with interleukin-1beta and a physiologically relevant concentration of KA (e.g., 100nM) leads to induction of IL6 expression that is largely dependent on AHR expression. Our findings have established that KA is a potent AHR endogenous ligand that can induce IL6 production and xenobiotic metabolism in cells at physiologically relevant concentrations.


Subject(s)
Excitatory Amino Acid Antagonists/metabolism , Hepatocytes/metabolism , Interleukin-6/biosynthesis , Kynurenic Acid/metabolism , Receptors, Aryl Hydrocarbon/agonists , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Environmental Pollutants/toxicity , Enzyme Induction/drug effects , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/pharmacology , Female , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenic Acid/chemistry , Kynurenic Acid/pharmacology , Ligands , Mice , Polychlorinated Dibenzodioxins/toxicity , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Structure-Activity Relationship
19.
Toxicol Sci ; 114(2): 217-25, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20044593

ABSTRACT

The human aryl hydrocarbon receptor (hAHR) and mouse aryl hydrocarbon receptor (mAHR(b)) share limited (58%) transactivation domain (TAD) sequence identity. Compared to the mAHR(b) allele, the hAHR displays 10-fold lower relative affinity for prototypical ligands, such as 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD). However, in previous studies, we have demonstrated that the hAHR can display a higher relative ligand-binding affinity than the mAHR(b) for specific AHR ligands, such as indirubin. Each receptor has also been shown to differentially recruit LXXLL coactivator motif proteins and to utilize different TAD subdomains in gene transactivation. Using hepatocytes isolated from C57BL/6J mice (Ahr(b/b)) and AHR(Ttr) transgenic mice, which express hAHR protein specifically in hepatocytes, we investigated whether the hAHR and mAHR(b) differentially regulate genes. DNA microarray and quantitative PCR analysis of Ahr(b/b) and AHR(Ttr) primary mouse hepatocytes treated with 10nM TCDD revealed that a number of established AHR target genes such as Cyp1a1 and Cyp1b1 are significantly induced by both receptors. Remarkably, of the 1752 genes induced by mAHR(b) and 1186 genes induced by hAHR, only 265 genes (approximately 18%) were significantly activated by both receptors in response to TCDD. Conversely, of the 1100 and 779 genes significantly repressed in mAHR(b) and hAHR hepatocytes, respectively, only 462 (approximately 49%) genes were significantly repressed by both receptors in response to TCDD treatment. Genes identified as differentially expressed are known to be involved in a number of biological pathways, including cell proliferation and inflammatory response, which suggest that compared to the mAHR(b), the hAHR may play contrasting roles in TCDD-induced toxicity and endogenous AHR-mediated gene regulation.


Subject(s)
Gene Expression Regulation/genetics , Receptors, Aryl Hydrocarbon/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Proliferation/drug effects , Environmental Pollutants/toxicity , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inflammation/chemically induced , Inflammation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/drug effects , Species Specificity
20.
Mol Pharmacol ; 77(2): 247-54, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19903824

ABSTRACT

The concept of selective receptor modulators has been established for the nuclear steroid hormone receptors. Such selective modulators have been used therapeutically with great success in the treatment of cancer. However, this concept has not been examined with regard to the aryl hydrocarbon receptor (AHR) because of the latent toxicity commonly associated with AHR activation. AHR-mediated toxicity is primarily derived from AHR binding to its dioxin response element (DRE) and driving expression of CYP1 family members, which have the capacity to metabolize procarcinogens to genotoxic carcinogens. Recent evidence using a non-DRE binding AHR mutant has established the DRE-independent suppression of inflammatory markers by the AHR. We wished to determine whether such DRE-independent repression with wild-type AHR could be dissociated from canonical DRE-dependent transactivation in a ligand-dependent manner and, in doing so, prove the concept of a selective AHR modulator (SAhRM). Here, we identify the selective estrogen receptor (ER) modulator Way-169916 as a dually selective modulator, binding both ER and AHR. Inflammatory gene expression associated with the cytokine-inducible acute-phase response (e.g., SAA1 and CRP) are diminished by Way-169916 in an AHR-dependent manner. Furthermore, activation of AHR by Way-169916 fails to stimulate canonical DRE-driven AHR-mediated CYP1A1 expression, thus eliminating the potential for AHR-mediated genotoxic stress. Such anti-inflammatory activity in the absence of DRE-mediated expression fulfills the major criteria of an SAhRM, which suggests that selective modulation of AHR is possible and renders the AHR a therapeutically viable drug target for the amelioration of inflammatory disease.


Subject(s)
Pyrazoles/chemistry , Pyrazoles/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Binding, Competitive/physiology , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Humans , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polychlorinated Dibenzodioxins/chemistry , Polychlorinated Dibenzodioxins/metabolism , Polychlorinated Dibenzodioxins/pharmacology , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...