193 results on '"Burris TP"'
Search Results
2. Merging Natural Product Structures with Pharmaceutical Leads: Unnatural Enantiomers of Estranes as Glucocorticoid Receptor Modulators That Suppress TNF-α and IL-6 Release.
- Author
-
Nicholson JM, Yang D, Koelblen T, Hu EL, Coss CC, Burris TP, Hu X, and Micalizio GC
- Subjects
- Stereoisomerism, Humans, Animals, Structure-Activity Relationship, Mice, Anti-Inflammatory Agents pharmacology, Anti-Inflammatory Agents chemistry, Receptors, Glucocorticoid metabolism, Receptors, Glucocorticoid agonists, Receptors, Glucocorticoid antagonists & inhibitors, Biological Products chemistry, Biological Products pharmacology, Tumor Necrosis Factor-alpha metabolism, Tumor Necrosis Factor-alpha antagonists & inhibitors, Interleukin-6 metabolism, Interleukin-6 antagonists & inhibitors
- Abstract
Natural products are widely recognized as valuable starting points for the development of therapeutics, with synthetic tetracyclic triterpenoids (e.g., steroids) being the most well represented among the drugs approved by the Food and Drug Administration. Here, recently developed synthetic tools for concise, asymmetric, and convergent construction of steroidal systems are leveraged to drive a program aimed at identifying novel glucocorticoid receptor (GR) modulators. While glucocorticoids have been extensively used as anti-inflammatory agents, they are plagued by severe side effects that include bone loss, muscle wasting, and metabolic disease. Ultimately, a program targeting the unnatural enantiomers of estranes ( ent -estranes) that are practically inaccessible from natural product derivatization (semisynthesis) has resulted in the identification of a new class of potent dissociated GR modulators. We identify several leads with >99% efficacy as antagonists of GR trans-activation (potency within 10-fold of that of mifepristone) and further characterize examples that also inhibit release of pro-inflammatory cytokines IL-6 and TNF-α.
- Published
- 2024
- Full Text
- View/download PDF
3. The Estrogen Receptor-Related Orphan Receptors Regulate Autophagy through TFEB.
- Author
-
Losby M, Hayes M, Valfort A, Sopariwala DH, Sanders R, Walker JK, Xu W, Narkar VA, Zhang L, Billon C, and Burris TP
- Subjects
- Animals, Rats, Mice, Humans, Cell Line, ERRalpha Estrogen-Related Receptor, Rats, Sprague-Dawley, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Autophagy physiology, Receptors, Estrogen metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac drug effects
- Abstract
Autophagy is an essential self-degradative and recycling mechanism that maintains cellular homeostasis. Estrogen receptor-related orphan receptors (ERRs) are fundamental in regulating cardiac metabolism and function. Previously, we showed that ERR agonists improve cardiac function in models of heart failure and induce autophagy. Here, we characterized a mechanism by which ERRs induce the autophagy pathway in cardiomyocytes. Transcription factor EB (TFEB) is a master regulator of the autophagy-lysosome pathway and has been shown to be crucial regulator of genes that control autophagy. We discovered that TFEB is a direct ERR target gene whose expression is induced by ERR agonists. Activation of ERR results in increased TFEB expression in both neonatal rat ventricular myocytes and C
2 C12 myoblasts. An ERR-dependent increase in TFEB expression results in increased expression of an array of TFEB target genes, which are critical for the stimulation of autophagy. Pharmacologically targeting ERR is a promising potential method for the treatment of many diseases where stimulation of autophagy may be therapeutic, including heart failure. SIGNIFICANCE STATEMENT: Estrogen receptor-related receptor agonists function as exercise mimetics and also display efficacy in animal models of metabolic disease, obesity, and heart failure., (Copyright © 2024 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2024
- Full Text
- View/download PDF
4. How to Make Glucocorticoids Safer.
- Author
-
Burris TP
- Subjects
- Glucocorticoids adverse effects
- Published
- 2024
- Full Text
- View/download PDF
5. From Functional Fatty Acids to Potent and Selective Natural-Product-Inspired Mimetics via Conformational Profiling.
- Author
-
Markham LE, Koelblen T, Chobanian HR, Follis AV, Burris TP, and Micalizio GC
- Abstract
Fatty acids play important signaling roles in biology, albeit typically lacking potency or selectivity, due to their substantial conformational flexibility. While being recognized as having properties of potentially great value as therapeutics, it is often the case that the functionally relevant conformation of the natural fatty acid is not known, thereby complicating efforts to develop natural-product-inspired ligands that have similar functional properties along with enhanced potency and selectivity profiles. In other words, without structural information associated with a particular functional relationship and the hopelessly unbiased conformational preferences of the endogenous ligand, one is molecularly ill-informed regarding the precise ligand-receptor interactions that play a role in driving the biological activity of interest. To address this problem, a molecular strategy to query the relevance of distinct subpopulations of fatty acid conformers has been established through "conformational profiling", a process whereby a unique collection of chiral and conformationally constrained fatty acids is employed to deconvolute beneficial structural features that impart natural-product-inspired function. Using oleic acid as an example because it is known to engage a variety of receptors, including GPR40, GPR120, and TLX, a 24-membered collection of mimetics was designed and synthesized. It was then demonstrated that this collection contained members that have enhanced potency and selectivity profiles, with some being clearly biased for engagement of the GPCRs GPR40 and GPR120 while others were identified as potent and selective modulators of the nuclear receptor TLX. A chemical synthesis strategy that exploited the power of modern technology for stereoselective synthesis was critical to achieving success, establishing a common sequence of bond-forming reactions to access a disparate collection of chiral mimetics, whose conformational preferences are impacted by the nature of stereodefined moieties differentially positioned about the C
18 skeleton of the parent fatty acid. Overall, this study establishes a foundation to fuel future programs aimed at developing natural-product-inspired fatty acid mimetics as valuable tools in chemical biology and potential therapeutic leads., Competing Interests: The authors declare no competing financial interest., (© 2024 The Authors. Published by American Chemical Society.)- Published
- 2024
- Full Text
- View/download PDF
6. A Synthetic ERR Agonist Alleviates Metabolic Syndrome.
- Author
-
Billon C, Schoepke E, Avdagic A, Chatterjee A, Butler AA, Elgendy B, Walker JK, and Burris TP
- Subjects
- Mice, Animals, Obesity drug therapy, Obesity metabolism, Energy Metabolism, Receptors, Cytoplasmic and Nuclear, ERRalpha Estrogen-Related Receptor, Estrogens, Metabolic Syndrome drug therapy, Insulin Resistance
- Abstract
Physical exercise induces physiologic adaptations and is effective at reducing the risk of premature death from all causes. Pharmacological exercise mimetics may be effective in the treatment of a range of diseases including obesity and metabolic syndrome. Previously, we described the development of SLU-PP-332, an agonist for the estrogen-related receptor (ERR) α , β, and γ nuclear receptors that activates an acute aerobic exercise program. Here we examine the effects of this exercise mimetic in mouse models of obesity and metabolic syndrome. Diet-induced obese or ob/ob mice were administered SLU-PP-332, and the effects on a range of metabolic parameters were assessed. SLU-PP-332 administration mimics exercise-induced benefits on whole-body metabolism in mice including increased energy expenditure and fatty acid oxidation. These effects were accompanied by decreased fat mass accumulation. Additionally, the ERR agonist effectively reduced obesity and improved insulin sensitivity in models of metabolic syndrome. Pharmacological activation of ERR may be an effective method to treat metabolic syndrome and obesity. SIGNIFICANCE STATEMENT: An estrogen receptor-related orphan receptor agonist, SLU-PP-332, with exercise mimetic activity, holds promise as a therapeutic to treat metabolic diseases by decreasing fat mass in mouse models of obesity., (Copyright © 2024 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2024
- Full Text
- View/download PDF
7. Estrogen-Related Receptor Agonism Reverses Mitochondrial Dysfunction and Inflammation in the Aging Kidney.
- Author
-
Wang XX, Myakala K, Libby AE, Krawczyk E, Panov J, Jones BA, Bhasin K, Shults N, Qi Y, Krausz KW, Zerfas PM, Takahashi S, Daneshpajouhnejad P, Titievsky A, Taranenko E, Billon C, Chatterjee A, Elgendy B, Walker JK, Albanese C, Kopp JB, Rosenberg AZ, Gonzalez FJ, Guha U, Brodsky L, Burris TP, and Levi M
- Subjects
- Mice, Humans, Animals, Aged, Infant, Infant, Newborn, Estrogens metabolism, Mitochondria metabolism, Cytokines metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Kidney metabolism, Inflammation metabolism
- Abstract
A gradual decline in renal function occurs even in healthy aging individuals. In addition to aging, per se, concurrent metabolic syndrome and hypertension, which are common in the aging population, can induce mitochondrial dysfunction and inflammation, which collectively contribute to age-related kidney dysfunction and disease. This study examined the role of the nuclear hormone receptors, the estrogen-related receptors (ERRs), in regulation of age-related mitochondrial dysfunction and inflammation. The ERRs were decreased in both aging human and mouse kidneys and were preserved in aging mice with lifelong caloric restriction (CR). A pan-ERR agonist, SLU-PP-332, was used to treat 21-month-old mice for 8 weeks. In addition, 21-month-old mice were treated with a stimulator of interferon genes (STING) inhibitor, C-176, for 3 weeks. Remarkably, similar to CR, an 8-week treatment with a pan-ERR agonist reversed the age-related increases in albuminuria, podocyte loss, mitochondrial dysfunction, and inflammatory cytokines, via the cyclic GMP-AMP synthase-STING and STAT3 signaling pathways. A 3-week treatment of 21-month-old mice with a STING inhibitor reversed the increases in inflammatory cytokines and the senescence marker, p21/cyclin dependent kinase inhibitor 1A (Cdkn1a), but also unexpectedly reversed the age-related decreases in PPARG coactivator (PGC)-1α, ERRα, mitochondrial complexes, and medium chain acyl coenzyme A dehydrogenase (MCAD) expression. These studies identified ERRs as CR mimetics and as important modulators of age-related mitochondrial dysfunction and inflammation. These findings highlight novel druggable pathways that can be further evaluated to prevent progression of age-related kidney disease., Competing Interests: Disclosure Statement None declared., (Copyright © 2023 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. International Union of Basic and Clinical Pharmacology CXIII: Nuclear Receptor Superfamily-Update 2023.
- Author
-
Burris TP, de Vera IMS, Cote I, Flaveny CA, Wanninayake US, Chatterjee A, Walker JK, Steinauer N, Zhang J, Coons LA, Korach KS, Cain DW, Hollenberg AN, Webb P, Forrest D, Jetten AM, Edwards DP, Grimm SL, Hartig S, Lange CA, Richer JK, Sartorius CA, Tetel M, Billon C, Elgendy B, Hegazy L, Griffett K, Peinetti N, Burnstein KL, Hughes TS, Sitaula S, Stayrook KR, Culver A, Murray MH, Finck BN, and Cidlowski JA
- Subjects
- Humans, Receptors, Cytoplasmic and Nuclear metabolism, Transcription Factors metabolism, Carrier Proteins, Ligands, Pharmacology, Clinical
- 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., (U.S. Government work not protected by U.S. copyright.)
- Published
- 2023
- Full Text
- View/download PDF
9. Development and pharmacological evaluation of a new chemical series of potent pan-ERR agonists, identification of SLU-PP-915.
- Author
-
Hampton CS, Sitaula S, Billon C, Haynes K, Avdagic A, Wanninayake U, Adeyemi CM, Chatterjee A, Griffett K, Banerjee S, Burris SL, Schoepke E, Boehm T, Bess A, de Vera IMS, Burris TP, and Walker JK
- Subjects
- Protein Isoforms, Estrogens
- Abstract
Estrogen-related receptors (ERR) are an orphan nuclear receptor sub-family that play a critical role in regulating gene transcription for several physiological processes including mitochondrial function, cellular energy utilization and homeostasis. They have also been implicated to play a role in several pathological conditions. Herein, we report the identification, synthesis, structure-activity relationships and pharmacological evaluation of a new chemical series of potent pan-ERR agonists. This template was designed for ERRγ starting from the known acyl hydrazide template and compounds such as agonist GSK-4716 employing a structure-based drug design approach. This led to the preparation of a series of 2,5-disubstituted thiophenes from which several were found to be potent agonists of ERRγ in cell-based co-transfection assays. Additionally, direct binding to ERRγ was established through
1 H NMR protein-ligand binding experiments. Compound optimization revealed that the phenolic or aniline groups could be replaced with a boronic acid moiety, which was able to maintain activity and demonstrated improved metabolic stability in microsomal in vitro assays. Further pharmacological evaluation of these compounds showed that they had roughly equivalent agonist activity on ERR isoforms α and β representing an ERR pan-agonist profile. One potent agonist, SLU-PP-915 (10s), which contained a boronic acid moiety was profiled in gene expression assays and found to significantly upregulate the expression of ERR target genes such as peroxisome-proliferator activated receptor γ co-activators-1α, lactate dehydrogenase A, DNA damage inducible transcript 4 and pyruvate dehydrogenase kinase 4 both in vitro and in vivo., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:John K Walker reports financial support was provided by National Institutes of Health. Thomas P. Burris reports financial support was provided by National Institutes of Health. John K. Walker has patent #WO2019036562; Heteroaromatics as ERR inverse agonists and their preparation licensed to Myonid Therapeutics. Thomas P. Burris has patent #WO2019036562; Heteroaromatics as ERR inverse agonists and their preparation licensed to Myonid Therapeutics. Carissa Hampton has patent #WO2019036562; Heteroaromatics as ERR inverse agonists and their preparation licensed to Myonid Therapeutics. Keith Haynes has patent #WO2019036562; Heteroaromatics as ERR inverse agonists and their preparation licensed to Myonid Therapeutics. Kristine Griffett has patent #WO2019036562; Heteroaromatics as ERR inverse agonists and their preparation licensed to Myonid Therapeutics. Cyrielle Billon has patent #WO2019036562; Heteroaromatics as ERR inverse agonists and their preparation licensed to Myonid Therapeutics. Sadichha Situala has patent #WO2019036562; Heteroaromatics as ERR inverse agonists and their preparation licensed to Myonid Therapeutics. Founders in Myonid Therapeutics which is trying to develop ERR modulators as novel therapeutics (J.K.W. & T.P.B), (Copyright © 2023 Elsevier Masson SAS. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
10. Rev-erbα agonists suppresses TGFβ1-induced fibroblast-to-myofibroblast transition and pro-fibrotic phenotype in human lung fibroblasts.
- Author
-
Prasad C, Hahn K, Duraisamy SK, Salathe MA, Huang SK, Burris TP, and Sundar IK
- Subjects
- Humans, Interleukin-6 metabolism, Lung pathology, Fibrosis, Fibroblasts metabolism, Phenotype, Chronic Disease, Nuclear Receptor Subfamily 1, Group D, Member 1 genetics, Nuclear Receptor Subfamily 1, Group D, Member 1 metabolism, Myofibroblasts metabolism, Idiopathic Pulmonary Fibrosis pathology
- Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by excessive scarring of the lungs that can lead to respiratory failure and death. Lungs of patients with IPF demonstrate excessive deposition of extracellular matrix (ECM) and an increased presence of pro-fibrotic mediators such as transforming growth factor-beta 1 (TGFβ1), which is a major driver of fibroblast-to-myofibroblast transition (FMT). Current literature supports that circadian clock dysfunction plays an essential role in the pathophysiology of various chronic inflammatory lung diseases such as asthma, chronic obstructive pulmonary disease, and IPF. The circadian clock transcription factor Rev-erbα is encoded by Nr1d1 that regulates daily rhythms of gene expression linked to immunity, inflammation, and metabolism. However, investigations into the potential roles of Rev-erbα in TGFβ-induced FMT and ECM accumulation are limited. In this study, we utilized several novel small molecule Rev-erbα agonists (GSK41122, SR9009, and SR9011) and a Rev-erbα antagonist (SR8278) to determine the roles of Rev-erbα in regulating TGFβ1-induced FMT and pro-fibrotic phenotypes in human lung fibroblasts. WI-38 cells were either pre-treated/co-treated with or without Rev-erbα agonist/antagonist along with TGFβ1. After 48 h, the following parameters were evaluated: secretion of COL1A1 (Slot-Blot analysis) and IL-6 (ELISA) into condition media, expressions of α-smooth muscle actin (αSMA: immunostaining and confocal microscopy), and pro-fibrotic proteins (αSMA and COL1A1 by immunoblotting), as well as gene expression of pro-fibrotic targets (qRT-PCR: Acta2, Fn1, and Col1a1). Results revealed that Rev-erbα agonists inhibited TGFβ1-induced FMT (αSMA and COL1A1), and ECM production (reduced gene expression of Acta2, Fn1, and Col1a1), and decreased pro-inflammatory cytokine IL-6 release. The Rev-erbα antagonist promoted TGFβ1-induced pro-fibrotic phenotypes. These findings support the potential of novel circadian clock-based therapeutics, such as Rev-erbα agonist, for the treatment and management of fibrotic lung diseases and disorders., Competing Interests: Declaration of competing interest The authors report no declarations of interest., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Synthetic ERRα/β/γ Agonist Induces an ERRα-Dependent Acute Aerobic Exercise Response and Enhances Exercise Capacity.
- Author
-
Billon C, Sitaula S, Banerjee S, Welch R, Elgendy B, Hegazy L, Oh TG, Kazantzis M, Chatterjee A, Chrivia J, Hayes ME, Xu W, Hamilton A, Huss JM, Zhang L, Walker JK, Downes M, Evans RM, and Burris TP
- Subjects
- Animals, Mice, Muscle Fibers, Skeletal metabolism, Muscle, Skeletal metabolism, ERRalpha Estrogen-Related Receptor, Exercise Tolerance drug effects, Receptors, Estrogen drug effects, Receptors, Estrogen metabolism, Estrogens chemistry, Estrogens pharmacology
- Abstract
Repetitive physical exercise induces physiological adaptations in skeletal muscle that improves exercise performance and is effective for the prevention and treatment of several diseases. Genetic evidence indicates that the orphan nuclear receptors estrogen receptor-related receptors (ERRs) play an important role in skeletal muscle exercise capacity. Three ERR subtypes exist (ERRα, β, and γ), and although ERRβ/γ agonists have been designed, there have been significant difficulties in designing compounds with ERRα agonist activity. Additionally, there are limited synthetic agonists that can be used to target ERRs in vivo . Here, we report the identification of a synthetic ERR pan agonist, SLU-PP-332, that targets all three ERRs but has the highest potency for ERRα. Additionally, SLU-PP-332 has sufficient pharmacokinetic properties to be used as an in vivo chemical tool. SLU-PP-332 increases mitochondrial function and cellular respiration in a skeletal muscle cell line. When administered to mice, SLU-PP-332 increased the type IIa oxidative skeletal muscle fibers and enhanced exercise endurance. We also observed that SLU-PP-332 induced an ERRα-specific acute aerobic exercise genetic program, and the ERRα activation was critical for enhancing exercise endurance in mice. These data indicate the feasibility of targeting ERRα for the development of compounds that act as exercise mimetics that may be effective in the treatment of numerous metabolic disorders and to improve muscle function in the aging.
- Published
- 2023
- Full Text
- View/download PDF
12. REV-ERB activation as a novel pharmacological approach for treating inflammatory pain.
- Author
-
Makhija S, Griffett JD, Veerakanellore GB, Burris TP, Elgendy B, and Griffett K
- Abstract
Pain is a complex problem affecting millions of people worldwide. The current therapies to reduce pain are limited as many treatment options inadequately address the causes of pain, lead to tolerance of the drug, or have adverse effects including abuse potential. While there are many causes of pain, one underlying mechanism to the pathogenesis and maintenance of pain conditions is chronic inflammation driven by the NLRP3 inflammasome. Several inflammasome inhibitors are currently under investigation however have the potential to suppress the functioning of the innate immune system, which may cause unwanted affects in patients. Here, we show that the nuclear receptor REV-ERB can suppress the activation of the inflammasome when pharmacologically activated with small molecule agonists. Additionally, REV-ERB activation appears to have analgesic potential in a model of acute inflammatory pain, likely as a result of inflammasome suppression., Competing Interests: TPB and BE hold stock in Pelagos Pharmaceuticals, Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Makhija, Griffett, Veerakanellore, Burris, Elgendy and Griffett.)
- Published
- 2023
- Full Text
- View/download PDF
13. Development of LXR inverse agonists to treat MAFLD, NASH, and other metabolic diseases.
- Author
-
Griffett K and Burris TP
- Abstract
Activation of LXR activity by synthetic agonists has been the focus of many drug discovery efforts with a focus on treatment of dyslipidemia and atherosclerosis. Many agonists have been developed, but all have been hindered due to their ability to efficaciously stimulate de novo lipogenesis. Here, we review the development of LXR inverse agonists that were originally optimized for their ability to enable recruitment of corepressors leading to silencing of genes that drive de novo lipogenesis. Such compounds have efficacy in animal models of MAFLD, dyslipidemia, and cancer. Several classes of LXR inverse agonists have been identified and one is now in clinical trials for treatment of severe dyslipidemia., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Griffett and Burris.)
- Published
- 2023
- Full Text
- View/download PDF
14. Structural basis of synthetic agonist activation of the nuclear receptor REV-ERB.
- Author
-
Murray MH, Valfort AC, Koelblen T, Ronin C, Ciesielski F, Chatterjee A, Veerakanellore GB, Elgendy B, Walker JK, Hegazy L, and Burris TP
- Subjects
- Heme metabolism, Ligands, Nuclear Receptor Subfamily 1, Group D, Member 1 genetics, Porphyrins pharmacology
- Abstract
The nuclear receptor REV-ERB plays an important role in a range of physiological processes. REV-ERB behaves as a ligand-dependent transcriptional repressor and heme has been identified as a physiological agonist. Our current understanding of how ligands bind to and regulate transcriptional repression by REV-ERB is based on the structure of heme bound to REV-ERB. However, porphyrin (heme) analogues have been avoided as a source of synthetic agonists due to the wide range of heme binding proteins and potential pleotropic effects. How non-porphyrin synthetic agonists bind to and regulate REV-ERB has not yet been defined. Here, we characterize a high affinity synthetic REV-ERB agonist, STL1267, and describe its mechanism of binding to REV-ERB as well as the method by which it recruits transcriptional corepressor both of which are unique and distinct from that of heme-bound REV-ERB., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
15. SR9009 improves heart function after pressure overload independent of cardiac REV-ERB.
- Author
-
Li H, Song S, Tien CL, Qi L, Graves A, Nasiotis E, Burris TP, Zhao Y, Sun Z, and Zhang L
- Abstract
The core clock component REV-ERB is essential for heart function. Previous studies show that REV-ERB agonist SR9009 ameliorates heart remodeling in the pressure overload model with transverse aortic constriction (TAC). However, it is unknown whether SR9009 indeed works through cardiac REV-ERB, given that SR9009 might target other proteins and that REV-ERB in non-cardiac tissues might regulate cardiac functions indirectly. To address this question, we generated the REV-ERBα/β cardiac-specific double knockout mice (cDKO). We found that REV-ERB cardiac deficiency leads to profound dilated cardiac myopathy after TAC compared to wild-type (WT) control mice, confirming the critical role of REV-ERB in protecting against pressure overload. Interestingly, the cardioprotective effect of SR9009 against TAC retains in cDKO mice. In addition, SR9009 administered at the time points corresponding to the peak or trough of REV-ERB expression showed similar cardioprotective effects, suggesting the REV-ERB-independent mechanisms in SR9009-mediated post-TAC cardioprotection. These findings highlight that genetic deletion of REV-ERB in cardiomyocytes accelerates adverse cardiac remodeling in response to pressure overload and demonstrated the REV-ERB-independent cardioprotective effect of SR9009 upon pressure overload., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Li, Song, Tien, Qi, Graves, Nasiotis, Burris, Zhao, Sun and Zhang.)
- Published
- 2022
- Full Text
- View/download PDF
16. Progress toward the De Novo Asymmetric Synthesis of Euphanes.
- Author
-
Wai H, Koelblen T, Hayes ME, Burris TP, and Micalizio GC
- Subjects
- Alkylation, Oxidation-Reduction, Stereoisomerism, Acids, Alkenes
- Abstract
Progress toward an asymmetric synthesis of euphanes is described. A C14-desmethyl euphane system possessing five differentially substituted and electronically distinct alkenes has been prepared. The route employed is based on sequential metallacycle-mediated annulative cross-coupling, double asymmetric Brønsted acid mediated intramolecular Friedel-Crafts alkylation, and an oxidative rearrangement to establish the requisite C10 quaternary center. These studies have also led to the discovery of a novel euphane-based modulator of the Liver X Receptor.
- Published
- 2022
- Full Text
- View/download PDF
17. Antihyperlipidemic Activity of Gut-Restricted LXR Inverse Agonists.
- Author
-
Griffett K, Hayes M, Bedia-Diaz G, Appourchaux K, Sanders R, Boeckman MP, Koelblen T, Zhang J, Schulman IG, Elgendy B, and Burris TP
- Subjects
- Animals, Cholesterol metabolism, Cholesterol, LDL therapeutic use, Hypolipidemic Agents therapeutic use, Liver X Receptors, Mice, Mice, Inbred C57BL, Atherosclerosis drug therapy, Atherosclerosis metabolism, Orphan Nuclear Receptors agonists, Orphan Nuclear Receptors genetics, Orphan Nuclear Receptors metabolism
- Abstract
Hyperlipidemia and increased circulating cholesterol levels are associated with increased cardiovascular disease risk. The liver X receptors (LXRs) are regulators of de novo lipogenesis and cholesterol transport and have been validated as potential therapeutic targets for the treatment of atherosclerosis. However, efforts to develop LXR agonists to reduce cardiovascular diseases have failed due to poor clinical outcomes-associated increased hepatic lipogenesis and elevated low-density lipoprotein (LDL) cholesterol (C). Here, we report that LXR inverse agonists are effective in lowering plasma LDL cholesterol and triglycerides in several models of hyperlipidemia, including the Ldlr null mouse model of atherosclerosis. Mechanistic studies demonstrate that LXR directly regulates the expression of Soat2 enzyme in the intestine, which is directly responsible for the re-uptake or excretion of circulating lipids. Oral administration of a gut-specific LXR inverse agonist leads to reduction of Soat2 expression in the intestine and effectively lowers circulating LDL cholesterol and triglyceride levels without modulating LXR target genes in the periphery. In summary, our studies highlight the therapeutic potential of the gut-restricted molecules to treat hyperlipidemia and atherosclerosis through the intestinal LXR- Soat2 axis.
- Published
- 2022
- Full Text
- View/download PDF
18. The role of REV-ERB in NASH.
- Author
-
Griffett K, Hayes ME, Boeckman MP, and Burris TP
- Subjects
- Animals, Circadian Rhythm physiology, Heme metabolism, Ligands, Transcription Factors metabolism, Non-alcoholic Fatty Liver Disease drug therapy, Nuclear Receptor Subfamily 1, Group D, Member 1
- Abstract
REV-ERBs are atypical nuclear receptors as they function as ligand-regulated transcriptional repressors. The natural ligand for the REV-ERBs (REV-ERBα and REV-ERBβ) is heme, and heme-binding results in recruitment of transcriptional corepressor proteins such as N-CoR that mediates repression of REV-ERB target genes. These two receptors regulate a large range of physiological processes including several important in the pathophysiology of non-alcoholic steatohepatitis (NASH). These include carbohydrate and lipid metabolism as well as inflammatory pathways. A number of synthetic REV-ERB agonists have been developed as chemical tools and they show efficacy in animal models of NASH. Here, we will review the functions of REV-ERB with regard to their relevance to NASH as well as the potential to target REV-ERB for treatment of this disease., (© 2022. The Author(s), under exclusive licence to Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Chinese Pharmacological Society.)
- Published
- 2022
- Full Text
- View/download PDF
19. Emerging Role of Nuclear Receptors for the Treatment of NAFLD and NASH.
- Author
-
Welch RD, Billon C, Losby M, Bedia-Diaz G, Fang Y, Avdagic A, Elgendy B, Burris TP, and Griffett K
- Abstract
Non-alcoholic fatty liver (NAFLD) over the past years has become a metabolic pandemic linked to a collection of metabolic diseases. The nuclear receptors ERRs, REV-ERBs, RORs, FXR, PPARs, and LXR are master regulators of metabolism and liver physiology. The characterization of these nuclear receptors and their biology has promoted the development of synthetic ligands. The possibility of targeting these receptors to treat NAFLD is promising, as several compounds including Cilofexor, thiazolidinediones, and Saroglitazar are currently undergoing clinical trials. This review focuses on the latest development of the pharmacology of these metabolic nuclear receptors and how they may be utilized to treat NAFLD and subsequent comorbidities.
- Published
- 2022
- Full Text
- View/download PDF
20. Synthesis and structure activity relationship of the first class of LXR inverse agonists.
- Author
-
Elgendy B, Griffett K, Hegazy L, Di Fruscia P, Sample K, Schoepke E, Kamenecka TM, and Burris TP
- Subjects
- Dose-Response Relationship, Drug, HEK293 Cells, Heterocyclic Compounds, 4 or More Rings chemistry, Humans, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Sulfonamides chemistry, Heterocyclic Compounds, 4 or More Rings pharmacology, Liver X Receptors agonists, Sulfonamides pharmacology
- Abstract
Liver X Receptors (LXRs) are members of the nuclear receptor family, and they play significant role in lipid and cholesterol metabolism. Moreover, they are key regulators of several inflammatory pathways. Pharmacological modulation of LXRs holds great potential in treatment of metabolic diseases, neurodegenerative diseases, and cancer. We were the first group to identify LXR inverse agonists SR9238 (6) and SR9243 (7) and demonstrate their potential utility in treating liver diseases and cancer. Here, we present the results of structure-activity relationship (SAR) studies, based around SR9238 (6) and SR9243 (7). This study led to identification of 16, 17, 19, and 38, which were more potent inverse agonists than SR9238 (6) and SR9243 (7) and inhibited expression of the fatty acid synthase gene in DU145 cells. We previously demonstrated that inhibition of FASN is correlated to the anticancer activity of SR9243 (7) and this suggests that new inverse agonists have great potential as anticancer agents. We identified compounds with distinct selectivity toward both LXR isoforms, which can be excellent tools to study the pharmacology of both isoforms. We employed molecular dynamic (MD) simulations to better understand the molecular mechanism underlying inverse agonist activity and to guide our future design., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
21. In Science Journals.
- Author
-
Grocholski B, Stern P, Stajic J, Vignieri S, Burris TP, Osborne IS, Williams I, Suleymanov Y, Sugden AM, Jiang D, Szuromi P, Charneski CA, Wong W, Alderton G, Zahn LM, and Ash C
- Abstract
Highlights from the Science family of journals.
- Published
- 2021
- Full Text
- View/download PDF
22. Naturally Occurring Genetic Variants in the Oxytocin Receptor Alter Receptor Signaling Profiles.
- Author
-
Malik M, Ward MD, Fang Y, Porter JR, Zimmerman MI, Koelblen T, Roh M, Frolova AI, Burris TP, Bowman GR, Imoukhuede PI, and England SK
- Abstract
The hormone oxytocin is commonly administered during childbirth to initiate and strengthen uterine contractions and prevent postpartum hemorrhage. However, patients have wide variation in the oxytocin dose required for a clinical response. To begin to uncover the mechanisms underlying this variability, we screened the 11 most prevalent missense genetic variants in the oxytocin receptor ( OXTR ) gene. We found that five variants, V45L, P108A, L206V, V281M, and E339K, significantly altered oxytocin-induced Ca
2+ signaling or β-arrestin recruitment and proceeded to assess the effects of these variants on OXTR trafficking to the cell membrane, desensitization, and internalization. The variants P108A and L206V increased OXTR localization to the cell membrane, whereas V281M and E339K caused OXTR to be retained inside the cell. We examined how the variants altered the balance between OXTR activation and desensitization, which is critical for appropriate oxytocin dosing. The E339K variant impaired OXTR activation, internalization, and desensitization to roughly equal extents. In contrast, V281M decreased OXTR activation but had no effect on internalization and desensitization. V45L and P108A did not alter OXTR activation but did impair β-arrestin recruitment, internalization, and desensitization. Molecular dynamics simulations predicted that V45L and P108A prevent extension of the first intracellular loop of OXTR, thus inhibiting β-arrestin binding. Overall, our data suggest mechanisms by which OXTR genetic variants could alter clinical response to oxytocin., Competing Interests: The authors declare no competing financial interest., (© 2021 The Authors. Published by American Chemical Society.)- Published
- 2021
- Full Text
- View/download PDF
23. Restoration of the molecular clock is tumor suppressive in neuroblastoma.
- Author
-
Moreno-Smith M, Milazzo G, Tao L, Fekry B, Zhu B, Mohammad MA, Di Giacomo S, Borkar R, Reddy KRK, Capasso M, Vasudevan SA, Sumazin P, Hicks J, Putluri N, Perini G, Eckel-Mahan K, Burris TP, and Barbieri E
- Subjects
- ARNTL Transcription Factors metabolism, Animals, Antineoplastic Agents therapeutic use, Benzamides pharmacology, Cell Line, Tumor, Cell Survival physiology, Humans, Lipogenesis physiology, Mice, Promoter Regions, Genetic genetics, Xenograft Model Antitumor Assays, N-Myc Proto-Oncogene Protein metabolism, Neuroblastoma drug therapy, Neuroblastoma pathology, Nuclear Receptor Subfamily 1, Group F, Member 1 metabolism
- Abstract
MYCN activation is a hallmark of advanced neuroblastoma (NB) and a known master regulator of metabolic reprogramming, favoring NB adaptation to its microenvironment. We found that the expression of the main regulators of the molecular clock loops is profoundly disrupted in MYCN-amplified NB patients, and this disruption independently predicts poor clinical outcome. MYCN induces the expression of clock repressors and downregulates the one of clock activators by directly binding to their promoters. Ultimately, MYCN attenuates the molecular clock by suppressing BMAL1 expression and oscillation, thereby promoting cell survival. Reestablishment of the activity of the clock activator RORα via its genetic overexpression and its stimulation through the agonist SR1078, restores BMAL1 expression and oscillation, effectively blocks MYCN-mediated tumor growth and de novo lipogenesis, and sensitizes NB tumors to conventional chemotherapy. In conclusion, reactivation of RORα could serve as a therapeutic strategy for MYCN-amplified NBs by blocking the dysregulation of molecular clock and cell metabolism mediated by MYCN.
- Published
- 2021
- Full Text
- View/download PDF
24. A two-hit model of alcoholic liver disease that exhibits rapid, severe fibrosis.
- Author
-
Sengupta M, Abuirqeba S, Kameric A, Cecile-Valfort A, Chatterjee A, Griffett K, Burris TP, and Flaveny CA
- 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., Competing Interests: The authors have declared no competing interests exist
- Published
- 2021
- Full Text
- View/download PDF
25. Fructose Promotes Cytoprotection in Melanoma Tumors and Resistance to Immunotherapy.
- Author
-
Kuehm LM, Khojandi N, Piening A, Klevorn LE, Geraud SC, McLaughlin NR, Griffett K, Burris TP, Pyles KD, Nelson AM, Preuss ML, Bockerstett KA, Donlin MJ, McCommis KS, DiPaolo RJ, and Teague RM
- Subjects
- Animals, Antineoplastic Agents, Immunological therapeutic use, Carcinoma, Cell Line, Tumor, Female, Heme Oxygenase-1 metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Neoplasms drug therapy, Cytoprotection, Drug Resistance, Neoplasm, Fructose metabolism, Neoplasms metabolism, Tumor Escape
- Abstract
Checkpoint blockade immunotherapy relies on the empowerment of the immune system to fight cancer. Why some patients fail to achieve durable clinical responses is not well understood, but unique individual factors such as diet, obesity, and related metabolic syndrome could play a role. The link between obesity and patient outcomes remains controversial and has been mired by conflicting reports and limited mechanistic insight. We addressed this in a C57BL/6 mouse model of diet-induced obesity using a Western diet high in both fats and sugars. Obese mice bearing B16 melanoma or MC38 carcinoma tumors had impaired immune responses to immunotherapy and a reduced capacity to control tumor progression. Unexpectedly, these compromised therapeutic outcomes were independent of body mass and, instead, were directly attributed to dietary fructose. Melanoma tumors in mice on the high-fructose diet were resistant to immunotherapy and showed increased expression of the cytoprotective enzyme heme oxygenase-1 (HO-1). This increase in HO-1 protein was recapitulated in human A375 melanoma cells exposed to fructose in culture. Induced expression of HO-1 shielded tumor cells from immune-mediated killing and was critical for resistance to checkpoint blockade immunotherapy, which could be overcome in vivo using a small-molecule inhibitor of HO-1. This study reveals dietary fructose as a driver of tumor immune evasion, identifying HO-1 expression as a mechanism of resistance and a promising molecular target for combination cancer immunotherapy. See article by Khojandi et al., p. 214 ., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
26. The Orphan Nuclear Receptor TLX Is a Receptor for Synthetic and Natural Retinoids.
- Author
-
Griffett K, Bedia-Diaz G, Hegazy L, de Vera IMS, Wanninayake US, Billon C, Koelblen T, Wilhelm ML, and Burris TP
- Subjects
- Binding Sites drug effects, Biological Products chemical synthesis, Biological Products pharmacology, Cells, Cultured, Dose-Response Relationship, Drug, Humans, Ligands, Male, Molecular Dynamics Simulation, Molecular Structure, Orphan Nuclear Receptors, Receptors, Cytoplasmic and Nuclear agonists, Retinoids chemical synthesis, Retinoids pharmacology, Young Adult, Biological Products chemistry, Receptors, Cytoplasmic and Nuclear chemistry, Retinoids chemistry
- Abstract
TLX is an orphan nuclear receptor that plays a critical role in both embryonic and adult neurogenesis, as well in the pathogenesis of glioblastomas. TLX functions predominately as a transcriptional repressor, but no natural ligands or high-affinity synthetic ligands have been identified. Here, we describe the identification of natural and synthetic retinoids as functional ligands for TLX. We identified potent synthetic retinoids that directly bind to TLX and either activate or inhibit its transcriptional repressor activity. Furthermore, we identified all-trans and 11-cis retinaldehyde (retinal), retinoids that play an essential role in the visual cycle, as the preferential natural retinoids that bind to and modulate the function of TLX. Molecular dynamics simulations followed by mutational analysis provided insight into the molecular basis of retinoid binding to TLX. Our data support the validity of TLX as a target for development of therapeutics to treat cognitive disorders and/or glioblastomas., Competing Interests: Declaration of Interests Authors declare no competing interests., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
27. Weak Microbial Metabolites: a Treasure Trove for Using Biomimicry to Discover and Optimize Drugs.
- Author
-
Dvorak Z, Klapholz M, Burris TP, Willing BP, Gioiello A, Pellicciari R, Galli F, March J, O'Keefe SJ, Sartor RB, Kim CH, Levy M, and Mani S
- Subjects
- Drug Discovery, Humans, Indoles chemistry, Ligands, Molecular Mimicry, Bacteria chemistry, Biological Products chemistry, Indoles pharmacology
- Abstract
For decades, traditional drug discovery has used natural product and synthetic chemistry approaches to generate libraries of compounds, with some ending as promising drug candidates. A complementary approach has been to adopt the concept of biomimicry of natural products and metabolites so as to improve multiple drug-like features of the parent molecule. In this effort, promiscuous and weak interactions between ligands and receptors are often ignored in a drug discovery process. In this Emerging Concepts article, we highlight microbial metabolite mimicry, whereby parent metabolites have weak interactions with their receptors that then have led to discrete examples of more potent and effective drug-like molecules. We show specific examples of parent-metabolite mimics with potent effects in vitro and in vivo. Furthermore, we show examples of emerging microbial ligand-receptor interactions and provide a context in which these ligands could be improved as potential drugs. A balanced conceptual advance is provided in which we also acknowledge potential pitfalls-hyperstimulation of finely balanced receptor-ligand interactions could also be detrimental. However, with balance, we provide examples of where this emerging concept needs to be tested. SIGNIFICANCE STATEMENT: Microbial metabolite mimicry is a novel way to expand on the chemical repertoire of future drugs. The emerging concept is now explained using specific examples of the discovery of therapeutic leads from microbial metabolites., Competing Interests: We (S.M., Z.D.) have filed a patent application US 2019/0367475 A1: PXR agonists and uses thereof for gut barrier dysfunction and treatment prevention., (Copyright © 2020 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2020
- Full Text
- View/download PDF
28. REV-ERB agonism improves liver pathology in a mouse model of NASH.
- Author
-
Griffett K, Bedia-Diaz G, Elgendy B, and Burris TP
- Subjects
- Animals, Disease Models, Animal, Humans, Inflammation metabolism, Lipogenesis physiology, Liver metabolism, Liver pathology, Liver Cirrhosis pathology, Mice, Obesity metabolism, Non-alcoholic Fatty Liver Disease drug therapy, Non-alcoholic Fatty Liver Disease pathology, Nuclear Receptor Subfamily 1, Group D, Member 1 drug effects, Pyrrolidines pharmacology, Thiophenes pharmacology
- Abstract
Non-alcoholic fatty liver disease (NAFLD) affects a significant number of people worldwide and currently there are no pharmacological treatments. NAFLD often presents with obesity, insulin resistance, and in some cases cardiovascular diseases. There is a clear need for treatment options to alleviate this disease since it often progresses to much more the much more severe non-alcoholic steatohepatitis (NASH). The REV-ERB nuclear receptor is a transcriptional repressor that regulates physiological processes involved in the development of NAFLD including lipogenesis and inflammation. We hypothesized that pharmacologically activating REV-ERB would suppress the progression of fatty liver in a mouse model of NASH. Using REV-ERB agonist SR9009 in a mouse NASH model, we demonstrate the beneficial effects of REV-ERB activation that led to an overall improvement of hepatic health by suppressing hepatic fibrosis and inflammatory response., Competing Interests: The authors have declared no competing interests.
- Published
- 2020
- Full Text
- View/download PDF
29. A Selective ERRα/γ Inverse Agonist, SLU-PP-1072, Inhibits the Warburg Effect and Induces Apoptosis in Prostate Cancer Cells.
- Author
-
Schoepke E, Billon C, Haynes KM, Avdagic A, Sitaula S, Sanders R, Adeyemi CM, Walker JK, and Burris TP
- Subjects
- Antineoplastic Agents chemical synthesis, Benzothiazoles chemical synthesis, Drug Inverse Agonism, Furans chemical synthesis, Gene Expression Regulation, Neoplastic drug effects, HEK293 Cells, Humans, PC-3 Cells, ERRalpha Estrogen-Related Receptor, Antineoplastic Agents pharmacology, Apoptosis drug effects, Benzothiazoles pharmacology, Furans pharmacology, Receptors, Estrogen antagonists & inhibitors, Warburg Effect, Oncologic drug effects
- Abstract
The estrogen related receptors (ERRs) are a subgroup of nuclear receptors that play a role in regulation of cellular metabolism. Prostate cancer (PCa) cells display altered metabolic signatures, such as the Warburg effect, and the ERRs have been implicated in driving this phenotype. Despite the lack of a known endogenous ligand, synthetic ligands that target the ERRs have been discovered. For example, the ERRα inverse agonist XCT790 modulates metabolic pathways in PCa cells, but it also functions as a mitochondrial uncoupler independent of targeting ERRα. Here, we describe a novel dual ERRα/γ inverse agonist, SLU-PP-1072, derived from the GSK4716 ERRγ agonist scaffold that is distinct from the XCT790 scaffold. SLU-PP-1072 alters PCa cell metabolism and gene expression, resulting in cell cycle dysregulation and increased apoptosis without acute mitochondrial uncoupling activity. Our data suggest that inhibition of ERRα/γ may be beneficial in treatment of PCa, and SLU-PP-1072 provides a unique chemical tool to evaluate the pharmacology of ERRα and ERRγ.
- Published
- 2020
- Full Text
- View/download PDF
30. Modulation of estrogen-related receptors subtype selectivity: Conversion of an ERRβ/γ selective agonist to ERRα/β/γ pan agonists.
- Author
-
Shahien M, Elagawany M, Sitaula S, Goher SS, Burris SL, Sanders R, Avdagic A, Billon C, Hegazy L, Burris TP, and Elgendy B
- Subjects
- Humans, Models, Molecular, Signal Transduction, Molecular Docking Simulation methods, Receptors, Estrogen metabolism
- Abstract
Estrogen Related Receptors (ERRs) are key regulators of energy homeostasis and play important role in the etiology of metabolic disorders, skeletal muscle related disorders, and neurodegenerative diseases. Among the three ERR isoforms, ERRα emerged as a potential drug target for metabolic and neurodegenerative diseases. Although ERRβ/γ selective agonist chemical tools have been identified, there are no chemical tools that effectively target ERRα agonism. We successfully engineered high affinity ERRα agonism into a chemical scaffold that displays selective ERRβ/γ agonist activity (GSK4716), providing novel ERRα/β/γ pan agonists that can be used as tools to probe the physiological roles of these nuclear receptors. We identified the structural requirements to enhance selectivity toward ERRα. Molecular modeling shows that our novel modulators have favorable binding modes in the LBP of ERRα and can induce conformational changes where Phe328 that originally occupies the pocket is dislocated to accommodate the ligands in a rather small cavity. The best agonists up-regulated the expression of target genes PGC-1α and PGC-1β, which are necessary to achieve maximal mitochondrial biogenesis. Moreover, they increased the mRNA levels of PDK4, which play an important role in energy homeostasis., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
31. Rev-erbα heterozygosity produces a dose-dependent phenotypic advantage in mice.
- Author
-
Welch RD, Billon C, Kameric A, Burris TP, and Flaveny CA
- Subjects
- 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, Dyslipidemias genetics, Muscle, Skeletal metabolism, Muscular Atrophy genetics, Nuclear Receptor Subfamily 1, Group D, Member 1 genetics
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
32. Clock regulation of protein secretion.
- Author
-
Burris TP
- Subjects
- Collagen, Homeostasis, CLOCK Proteins, Secretory Pathway
- Published
- 2020
- Full Text
- View/download PDF
33. Circadian rhythm-dependent and circadian rhythm-independent impacts of the molecular clock on type 3 innate lymphoid cells.
- Author
-
Wang Q, Robinette ML, Billon C, Collins PL, Bando JK, Fachi JL, Sécca C, Porter SI, Saini A, Gilfillan S, Solt LA, Musiek ES, Oltz EM, Burris TP, and Colonna M
- Subjects
- Animals, Mice, Mice, Congenic, Mice, Inbred C57BL, Mice, Knockout, Nuclear Receptor Subfamily 1, Group D, Member 1 deficiency, Nuclear Receptor Subfamily 1, Group D, Member 1 immunology, Circadian Rhythm immunology, Immunity, Innate immunology, Intestines immunology, Lymphocytes immunology, Nuclear Receptor Subfamily 1, Group F, Member 3 immunology
- Abstract
Many gut functions are attuned to circadian rhythm. Intestinal group 3 innate lymphoid cells (ILC3s) include NKp46
+ and NKp46- subsets, which are RORγt dependent and provide mucosal defense through secretion of interleukin-22 (IL-22) and IL-17. Because ILC3s highly express some key circadian clock genes, we investigated whether ILC3s are also attuned to circadian rhythm. We noted circadian oscillations in the expression of clock and cytokine genes, such as REV-ERBα, IL-22, and IL-17, whereas acute disruption of the circadian rhythm affected cytokine secretion by ILC3s. Because of prominent and rhythmic expression of REV-ERBα in ILC3s, we also investigated the impact of constitutive deletion of REV-ERBα, which has been previously shown to inhibit the expression of a RORγt repressor, NFIL3, while also directly antagonizing DNA binding of RORγt. Development of the NKp46+ ILC3 subset was markedly impaired, with reduced cell numbers, RORγt expression, and IL-22 production in REV-ERBα-deficient mice. The NKp46- ILC3 subsets developed normally, potentially due to compensatory expression of other clock genes, but IL-17 secretion paradoxically increased, probably because RORγt was not antagonized by REV-ERBα. We conclude that ILC3s are attuned to circadian rhythm, but clock regulator REV-ERBα also has circadian-independent impacts on ILC3 development and functions due to its roles in the regulation of RORγt., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
34. SR9009 administered for one day after myocardial ischemia-reperfusion prevents heart failure in mice by targeting the cardiac inflammasome.
- Author
-
Reitz CJ, Alibhai FJ, Khatua TN, Rasouli M, Bridle BW, Burris TP, and Martino TA
- Subjects
- Animals, Biomarkers, Biopsy, Heart Failure pathology, Heart Failure prevention & control, Immunohistochemistry, Mice, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Nuclear Receptor Subfamily 1, Group D, Member 1 genetics, Heart Failure etiology, Heart Failure metabolism, Inflammasomes metabolism, Myocardial Ischemia complications, Myocardial Ischemia metabolism, Myocardial Reperfusion, Pyrrolidines pharmacology, Thiophenes pharmacology
- Abstract
Reperfusion of patients after myocardial infarction (heart attack) triggers cardiac inflammation that leads to infarct expansion and heart failure (HF). We previously showed that the circadian mechanism is a critical regulator of reperfusion injury. However, whether pharmacological targeting using circadian medicine limits reperfusion injury and protects against HF is unknown. Here, we show that short-term targeting of the circadian driver REV-ERB with SR9009 benefits long-term cardiac repair post-myocardial ischemia reperfusion in mice. Gain and loss of function studies demonstrate specificity of targeting REV-ERB in mice. Treatment for just one day abates the cardiac NLRP3 inflammasome, decreasing immunocyte recruitment, and thereby allowing the vulnerable infarct to heal. Therapy is given in vivo, after reperfusion, and promotes efficient repair. This study presents downregulation of the cardiac inflammasome in fibroblasts as a cellular target of SR9009, inviting more targeted therapeutic investigations in the future., Competing Interests: Competing interestsThe authors declare no competing interests., (© The Author(s) 2019.)
- Published
- 2019
- Full Text
- View/download PDF
35. The nuclear receptor REV-ERBα modulates Th17 cell-mediated autoimmune disease.
- Author
-
Chang C, Loo CS, Zhao X, Solt LA, Liang Y, Bapat SP, Cho H, Kamenecka TM, Leblanc M, Atkins AR, Yu RT, Downes M, Burris TP, Evans RM, and Zheng Y
- Subjects
- Amino Acid Motifs genetics, Amino Acid Motifs immunology, Animals, Cell Differentiation genetics, Cell Differentiation immunology, Encephalomyelitis, Autoimmune, Experimental drug therapy, Encephalomyelitis, Autoimmune, Experimental genetics, Gene Expression Regulation drug effects, Gene Expression Regulation immunology, Genetic Loci, HEK293 Cells, Humans, Interleukin-17 genetics, Interleukin-17 immunology, Interleukin-17 metabolism, Mice, Mice, Transgenic, Multiple Sclerosis drug therapy, Multiple Sclerosis genetics, Nuclear Receptor Subfamily 1, Group D, Member 1 agonists, Nuclear Receptor Subfamily 1, Group D, Member 1 immunology, Nuclear Receptor Subfamily 1, Group F, Member 3 immunology, Pyrrolidines pharmacology, Pyrrolidines therapeutic use, RNA-Seq, Response Elements genetics, Th17 Cells metabolism, Thiophenes pharmacology, Thiophenes therapeutic use, Encephalomyelitis, Autoimmune, Experimental immunology, Multiple Sclerosis immunology, Nuclear Receptor Subfamily 1, Group D, Member 1 metabolism, Nuclear Receptor Subfamily 1, Group F, Member 3 metabolism, Th17 Cells immunology
- Abstract
T helper 17 (Th17) cells produce interleukin-17 (IL-17) cytokines and drive inflammatory responses in autoimmune diseases such as multiple sclerosis. The differentiation of Th17 cells is dependent on the retinoic acid receptor-related orphan nuclear receptor RORγt. Here, we identify REV-ERBα (encoded by Nr1d1 ), a member of the nuclear hormone receptor family, as a transcriptional repressor that antagonizes RORγt function in Th17 cells. REV-ERBα binds to ROR response elements (RORE) in Th17 cells and inhibits the expression of RORγt-dependent genes including Il17a and Il17f Furthermore, elevated REV-ERBα expression or treatment with a synthetic REV-ERB agonist significantly delays the onset and impedes the progression of experimental autoimmune encephalomyelitis (EAE). These results suggest that modulating REV-ERBα activity may be used to manipulate Th17 cells in autoimmune diseases., Competing Interests: The authors declare no conflict of interest., (Copyright © 2019 the Author(s). Published by PNAS.)
- Published
- 2019
- Full Text
- View/download PDF
36. A synthesis strategy for tetracyclic terpenoids leads to agonists of ERβ.
- Author
-
Kim WS, Shalit ZA, Nguyen SM, Schoepke E, Eastman A, Burris TP, Gaur AB, and Micalizio GC
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Survival drug effects, Drug Development, Humans, Neural Stem Cells drug effects, Stereoisomerism, Terpenes pharmacology, Brain Neoplasms, Estrogen Receptor beta agonists, Glioblastoma, Terpenes chemical synthesis
- Abstract
Natural product and natural product-like molecules continue to be important for the development of pharmaceutical agents, as molecules in this class play a vital role in the pipeline for new therapeutics. Among these, tetracyclic terpenoids are privileged, with >100 being FDA-approved drugs. Despite this significant pharmaceutical success, there remain considerable limitations to broad medicinal exploitation of the class due to lingering scientific challenges associated with compound availability. Here, we report a concise asymmetric route to forging natural and unnatural (enantiomeric) C19 and C20 tetracyclic terpenoid skeletons suitable to drive medicinal exploration. While efforts have been focused on establishing the chemical science, early investigations reveal that the emerging chemical technology can deliver compositions of matter that are potent and selective agonists of the estrogen receptor beta, and that are selectively cytotoxic in two different glioblastoma cell lines (U251 and U87).
- Published
- 2019
- Full Text
- View/download PDF
37. Pharmacological activation of the nuclear receptor REV-ERB reverses cognitive deficits and reduces amyloid-β burden in a mouse model of Alzheimer's disease.
- Author
-
Roby DA, Ruiz F, Kermath BA, Voorhees JR, Niehoff M, Zhang J, Morley JE, Musiek ES, Farr SA, and Burris TP
- Subjects
- Animals, Disease Models, Animal, Disks Large Homolog 4 Protein metabolism, Hippocampus metabolism, Hippocampus pathology, Male, Mice, Alzheimer Disease drug therapy, Alzheimer Disease metabolism, Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Cognitive Dysfunction drug therapy, Cognitive Dysfunction metabolism, Cognitive Dysfunction pathology, Nuclear Receptor Subfamily 1, Group D, Member 1 metabolism, Peptide Fragments metabolism, Pyrrolidines pharmacology, Thiophenes pharmacology
- Abstract
Alzheimer's disease currently lacks treatment options that effectively reverse the biological/anatomical pathology and cognitive deficits associated with the disease. Loss of function of the nuclear receptor REV-ERB is associated with reduced cognitive function in mouse models. The effect of enhanced REV-ERB activity on cognitive function has not been examined. In this study, we tested the hypothesis that enhanced REV-ERB function may enhance cognitive function in a model of Alzheimer's disease. We utilized the REV-ERB agonist SR9009 to pharmacologically activate the activity of REV-ERB in the SAMP8 mouse model of Alzheimer's disease. SR9009 reversed cognitive dysfunction of an aged SAMP8 mouse in several behavioral assays including novel object recognition, T-maze foot shock avoidance, and lever press operant conditioning task assessments. SR9009 treatment reduced amyloid-β 1-40 and 1-42 levels in the cortex, which is consistent with improved cognitive function. Furthermore, SR9009 treatment led to increased hippocampal PSD-95, cortical synaptophysin expression and the number of synapses suggesting improvement in synaptic function. We conclude that REV-ERB is a potential target for treatment of Alzheimer's disease., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
38. Circadian clock protein Rev-erbα regulates neuroinflammation.
- Author
-
Griffin P, Dimitry JM, Sheehan PW, Lananna BV, Guo C, Robinette ML, Hayes ME, Cedeño MR, Nadarajah CJ, Ezerskiy LA, Colonna M, Zhang J, Bauer AQ, Burris TP, and Musiek ES
- Subjects
- Animals, Astrocytes metabolism, Astrocytes pathology, Cell Death, Gene Deletion, Gliosis pathology, Hippocampus pathology, Lipopolysaccharides, Mice, Inbred C57BL, Mice, Knockout, Microglia metabolism, Microglia pathology, NF-kappa B metabolism, Nerve Net metabolism, Nuclear Receptor Subfamily 1, Group D, Member 1 deficiency, Signal Transduction, Circadian Clocks, Inflammation metabolism, Inflammation pathology, Neurons metabolism, Neurons pathology, Nuclear Receptor Subfamily 1, Group D, Member 1 metabolism
- Abstract
Circadian dysfunction is a common attribute of many neurodegenerative diseases, most of which are associated with neuroinflammation. Circadian rhythm dysfunction has been associated with inflammation in the periphery, but the role of the core clock in neuroinflammation remains poorly understood. Here we demonstrate that Rev-erbα, a nuclear receptor and circadian clock component, is a mediator of microglial activation and neuroinflammation. We observed time-of-day oscillation in microglial immunoreactivity in the hippocampus, which was disrupted in Rev-erbα
-/- mice. Rev-erbα deletion caused spontaneous microglial activation in the hippocampus and increased expression of proinflammatory transcripts, as well as secondary astrogliosis. Transcriptomic analysis of hippocampus from Rev-erbα-/- mice revealed a predominant inflammatory phenotype and suggested dysregulated NF-κB signaling. Primary Rev-erbα-/- microglia exhibited proinflammatory phenotypes and increased basal NF-κB activation. Chromatin immunoprecipitation revealed that Rev-erbα physically interacts with the promoter regions of several NF-κB-related genes in primary microglia. Loss of Rev-erbα in primary astrocytes had no effect on basal activation but did potentiate the inflammatory response to lipopolysaccharide (LPS). In vivo, Rev-erbα-/- mice exhibited enhanced hippocampal neuroinflammatory responses to peripheral LPS injection, while pharmacologic activation of Rev-erbs with the small molecule agonist SR9009 suppressed LPS-induced hippocampal neuroinflammation. Rev-erbα deletion influenced neuronal health, as conditioned media from Rev-erbα-deficient primary glial cultures exacerbated oxidative damage in cultured neurons. Rev-erbα-/- mice also exhibited significantly altered cortical resting-state functional connectivity, similar to that observed in neurodegenerative models. Our results reveal Rev-erbα as a pharmacologically accessible link between the circadian clock and neuroinflammation., Competing Interests: The authors declare no conflict of interest.- Published
- 2019
- Full Text
- View/download PDF
39. Development of novel liver X receptor modulators based on a 1,2,4-triazole scaffold.
- Author
-
Goher SS, Griffett K, Hegazy L, Elagawany M, Arief MMH, Avdagic A, Banerjee S, Burris TP, and Elgendy B
- Subjects
- Dose-Response Relationship, Drug, Drug Development, Humans, Liver X Receptors genetics, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Triazoles chemical synthesis, Triazoles chemistry, Liver X Receptors agonists, Triazoles pharmacology
- Abstract
Liver X Receptor (LXR) agonists have been reported as a potential treatment for atherosclerosis, Alzheimer's disease and hepatitis C virus (HCV) infection. We have designed and synthesized a series of potent compounds based on a 1,2,4-triazole scaffold as novel LXR modulators. In cell-based cotransfection assays these compounds generally functioned as LXR agonists and we observed compounds with selectivity towards LXRα (7-fold) and LXRβ (7-fold) in terms of potency. Assessment of the effects of selected compounds on LXR target gene expression in HepG2 cells revealed that compounds 6a-b and 8a-b behaved as inverse agonists on FASN expression even though they were agonists in the LXRα and LXRβ cotransfection assays. Interestingly, these compounds had no effect on the expression of SREBP-1c confirming a unique LXR modulator pharmacology. Molecular docking studies and evaluation of ADME properties in-silico show that active compounds possess favorable binding modes and ADME profiles. Thus, these compounds may be useful for in vivo characterization of LXR modulators with unique profiles and determination of their potential clinical utility., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
40. Doubled lifespan and patient-like pathologies in progeria mice fed high-fat diet.
- Author
-
Kreienkamp R, Billon C, Bedia-Diaz G, Albert CJ, Toth Z, Butler AA, McBride-Gagyi S, Ford DA, Baldan A, Burris TP, and Gonzalo S
- Subjects
- Animals, Diet, High-Fat, Humans, Lamin Type A genetics, Lamin Type A metabolism, Mice, Inbred C57BL, Mutation genetics, Phenotype, Progeria metabolism, Feeding Behavior, Longevity, Progeria pathology
- Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a devastating premature aging disease. Mouse models have been instrumental for understanding HGPS mechanisms and for testing therapies, which to date have had only marginal benefits in mice and patients. Barriers to developing effective therapies include the unknown etiology of progeria mice early death, seemingly unrelated to the reported atherosclerosis contributing to HGPS patient mortality, and mice not recapitulating the severity of human disease. Here, we show that progeria mice die from starvation and cachexia. Switching progeria mice approaching death from regular diet to high-fat diet (HFD) rescues early lethality and ameliorates morbidity. Critically, feeding the mice only HFD delays aging and nearly doubles lifespan, which is the greatest lifespan extension recorded in progeria mice. The extended lifespan allows for progeria mice to develop degenerative aging pathologies of a severity that emulates the human disease. We propose that starvation and cachexia greatly influence progeria phenotypes and that nutritional/nutraceutical strategies might help modulate disease progression. Importantly, progeria mice on HFD provide a more clinically relevant animal model to study mechanisms of HGPS pathology and to test therapies., (© 2018 The Authors. Aging Cell published by the Anatomical Society and John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
41. Corrigendum to "Pharmacological targeting of the mammalian clock reveals a novel analgesic for osteoarthritis-induced pain" [GENE 655 (2018) 1-12].
- Author
-
Das V, Kc R, Li X, Varma D, Qiu S, Kroin JS, Forsyth CB, Keshavarzian A, van Wijnen AJ, Park TJ, Stein GS, O-Sullivan I, Burris TP, and Im HJ
- Published
- 2019
- Full Text
- View/download PDF
42. RORγ regulates the NLRP3 inflammasome.
- Author
-
Billon C, Murray MH, Avdagic A, and Burris TP
- Subjects
- Animals, Galactosamine toxicity, Inflammasomes genetics, Interleukin-1beta genetics, Interleukin-1beta immunology, Lipopolysaccharides toxicity, Liver Failure, Acute chemically induced, Liver Failure, Acute genetics, Liver Failure, Acute immunology, Liver Failure, Acute pathology, Macrophages pathology, Mice, Mice, Knockout, NLR Family, Pyrin Domain-Containing 3 Protein genetics, Nuclear Receptor Subfamily 1, Group F, Member 3 genetics, Response Elements immunology, Sepsis chemically induced, Sepsis genetics, Sepsis immunology, Sepsis pathology, Th17 Cells pathology, Immunity, Innate, Inflammasomes immunology, Macrophages immunology, NLR Family, Pyrin Domain-Containing 3 Protein immunology, Nuclear Receptor Subfamily 1, Group F, Member 3 immunology, Th17 Cells immunology
- Abstract
RAR-related orphan receptor γ (RORγ) is a nuclear receptor that plays an essential role in the development of T helper 17 (T
h 17) cells of the adaptive immune system. The NLRP3 inflammasome is a component of the innate immune system that processes interleukin (IL)-1β into a mature cytokine. Elevated activity of the NLRP3 inflammasome contributes to the progression of an array of inflammatory diseases. Bone marrow-derived macrophages (BMDMs) isolated from RORγ-null mice displayed reduced capacity to secrete IL-1β, and they also displayed a reduction in Nlrp3 and Il1b gene expression. Examination of the promoters of the Il1b and Nlrp3 genes revealed multiple putative ROR response elements (ROREs) that were occupied by RORγ. RORγ inverse agonists were effective inhibitors of the inflammasome. RORγ inverse agonists suppressed lipopolysaccharide (LPS)/ATP-stimulated IL-1β secretion and expression of Il1b and Nlrp3 in BMDMs. Additionally, the ability of the RORγ inverse agonists to suppress IL-1β secretion was lost in Nlrp3 -null macrophages. The potential for targeting the NLRP3 inflammasome in vivo using RORγ inverse agonists was examined in two models: LPS-induced sepsis and fulminant hepatitis. Pharmacological inhibition of RORγ activity reduced plasma IL-1β as well as IL-1β production by peritoneal macrophages in a model of LPS-induced sepsis. Additionally, RORγ inverse agonists reduced mortality in an LPS/d-galactosamine-induced fulminant hepatitis mouse model. These results illustrate a major role for RORγ in regulation of innate immunity via modulation of NLRP3 inflammasome activity. Furthermore, these data suggest that inhibiting the NLRP3 inflammasome with RORγ inverse agonists may be an effective method to treat NLRP3-associated diseases., (© 2019 Billon et al.)- Published
- 2019
- Full Text
- View/download PDF
43. Recent Advances in the Medicinal Chemistry of Liver X Receptors.
- Author
-
El-Gendy BEM, Goher SS, Hegazy LS, Arief MMH, and Burris TP
- Subjects
- Animals, Atherosclerosis drug therapy, Cholesterol metabolism, Humans, Lipid Metabolism drug effects, Liver X Receptors chemistry, Liver X Receptors metabolism, Steroids chemistry, Steroids pharmacology, Structure-Activity Relationship, Liver X Receptors agonists, Liver X Receptors antagonists & inhibitors
- Abstract
Nuclear hormone receptors represent a large family of ligand-activated transcription factors that include steroid receptors, thyroid/retinoid receptors, and orphan receptors. Among nuclear hormone receptors, the liver X receptors have emerged as very important drug targets. These receptors regulate some of the most important metabolic functions, and they were also identified as anti-inflammatory transcription factors and regulators of the immune system. The development of drugs targeting liver X receptors continues to be a challenge, but advances in our knowledge of receptor structure and function move us forward, toward achieving this goal. This review highlights the latest advances in the development of synthetic LXR modulators in the primary literature from 2013 to 2017. In this review, we place great emphasis on the structure and function of LXRs because of their essential role in the drug design process. The structure-activity relationships of the most active and promising synthetic modulators are discussed.
- Published
- 2018
- Full Text
- View/download PDF
44. Inhibition of Hepatotoxicity by a LXR Inverse Agonist in a Model of Alcoholic Liver Disease.
- Author
-
Sengupta M, Griffett K, Flaveny CA, and Burris TP
- 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., Competing Interests: The authors declare no competing financial interest.
- Published
- 2018
- Full Text
- View/download PDF
45. Pharmacological targeting of the mammalian clock reveals a novel analgesic for osteoarthritis-induced pain.
- Author
-
Das V, Kc R, Li X, Varma D, Qiu S, Kroin JS, Forsyth CB, Keshavarzian A, van Wijnen AJ, Park TJ, Stein GS, O-Sullivan I, Burris TP, and Im HJ
- Subjects
- Animals, Arthralgia genetics, CLOCK Proteins genetics, Female, Hyperalgesia drug therapy, Hyperalgesia genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mutagenesis physiology, NAV1.8 Voltage-Gated Sodium Channel genetics, Osteoarthritis genetics, Analgesics therapeutic use, Arthralgia drug therapy, Circadian Rhythm drug effects, Circadian Rhythm genetics, Molecular Targeted Therapy methods, Osteoarthritis drug therapy
- Abstract
Environmental disruption of the circadian rhythm is linked with increased pain due to osteoarthritis (OA). We aimed to characterize the role of the clock gene in OA-induced pain more systemically using both genetic and pharmacological approaches. Genetically modified mice, (bmal1f/fNav1.8CreERT mice), generated by deleting the critical clock gene, bmal1, from Nav1.8 sensory neurons, were resistant to the development of mechanical hyperalgesia associated with OA induced by partial medial meniscectomy (PMM) of the knee. In wild-type mice, induction of OA by PMM surgery led to a substantial increase in BMAL1 expression in DRG neurons. Interestingly, pharmacological activation of the REV-ERB (a negative regulator of bmal1 transcription) with SR9009 resulted in reduction of BMAL1 expression, and a significant decrease in mechanical hyperalgesia associated with OA. Cartilage degeneration was also significantly reduced in mice treated with the REV-ERB agonist SR9009. Based on these data, we also assessed the effect of pharmacological activation of REV-ERB using a model of environmental circadian disruption with its associated mechanical hyperalgesia, and noted that SR9009 was an effective analgesic in this model as well. Our data clearly demonstrate that genetic disruption of the molecular clock, via deletion of bmal1 in the sensory neurons of the DRG, decreases pain in a model of OA. Furthermore, pharmacological activation of REV-ERB leading to suppression of BMAL1 expression may be an effective method for treating OA-related pain, as well as to reduce joint damage associated with this disease., (Copyright © 2018. Published by Elsevier B.V.)
- Published
- 2018
- Full Text
- View/download PDF
46. Distinct roles for REV-ERBα and REV-ERBβ in oxidative capacity and mitochondrial biogenesis in skeletal muscle.
- Author
-
Amador A, Campbell S, Kazantzis M, Lan G, Burris TP, and Solt LA
- Subjects
- Animals, Body Weight, Calorimetry, Indirect, Cell Line, Circadian Rhythm genetics, Energy Metabolism genetics, Fatty Acids metabolism, Feeding Behavior physiology, Female, Gene Expression Regulation, Male, Mice, Mice, Knockout, Nuclear Receptor Subfamily 1, Group D, Member 1 antagonists & inhibitors, Nuclear Receptor Subfamily 1, Group D, Member 1 deficiency, Nuclear Receptor Subfamily 1, Group D, Member 1 genetics, Organelle Biogenesis, Oxidation-Reduction, Oxidative Phosphorylation, RNA Interference, RNA, Small Interfering genetics, Receptors, Cytoplasmic and Nuclear antagonists & inhibitors, Receptors, Cytoplasmic and Nuclear deficiency, Receptors, Cytoplasmic and Nuclear genetics, Repressor Proteins antagonists & inhibitors, Repressor Proteins deficiency, Repressor Proteins genetics, Circadian Rhythm physiology, Energy Metabolism physiology, Mitochondria, Muscle physiology, Muscle, Skeletal metabolism, Nuclear Receptor Subfamily 1, Group D, Member 1 physiology, Receptors, Cytoplasmic and Nuclear physiology, Repressor Proteins physiology
- Abstract
The nuclear receptors REV-ERBα and REV-ERBβ have been demonstrated to be core members of the circadian clock and participate in the regulation of a diverse set of metabolic functions. Due to their overlapping tissue expression patterns and gene expression profiles, REV-ERBβ is thought to be redundant to REV-ERBα. Recent work has highlighted REV-ERBα's role in the regulation of skeletal muscle oxidative capacity and mitochondrial biogenesis. Considering the similarity between the REV-ERBs and the hypothesized overlap in function, we sought to determine whether REV-ERBβ-deficiency presented with a similar skeletal muscle phenotype as REV-ERBα-deficiency. Ectopic overexpression in C2C12 cells demonstrated that REV-ERBβ drives mitochondrial biogenesis and the expression of genes involved in fatty acid oxidation. Intriguingly, knock down of REV-ERBβ in C2C12 cultures also resulted in mitochondrial biogenesis and increased expression of genes involved in fatty acid β-oxidation. To determine whether these effects occurred in vivo, we examined REV-ERBβ-deficient mice and observed a similar increase in expression of genes involved in mitochondrial biogenesis and fatty acid β-oxidation. Consistent with these results, REV-ERBβ-deficient mice exhibited an altered metabolic phenotype compared to wild-type littermate controls when measured by indirect calorimetry. This likely compensated for the increased food consumption that occurred, possibly aiding in the maintenance of their weight over time. Since feeding behaviors are a direct circadian output, this study suggests that REV-ERBβ may have more subtle effects on circadian behaviors than originally identified. Furthermore, these data implicate REV-ERBβ in the control of skeletal muscle metabolism and energy expenditure and suggest that development of REV-ERBα versus REV-ERBβ selective ligands may have therapeutic utility in the treatment of metabolic syndrome.
- Published
- 2018
- Full Text
- View/download PDF
47. Nuclear Receptor Subfamily 1 Group D Member 1 Regulates Circadian Activity of NLRP3 Inflammasome to Reduce the Severity of Fulminant Hepatitis in Mice.
- Author
-
Pourcet B, Zecchin M, Ferri L, Beauchamp J, Sitaula S, Billon C, Delhaye S, Vanhoutte J, Mayeuf-Louchart A, Thorel Q, Haas JT, Eeckhoute J, Dombrowicz D, Duhem C, Boulinguiez A, Lancel S, Sebti Y, Burris TP, Staels B, and Duez HM
- Subjects
- Animals, Caspase 1 metabolism, Cells, Cultured, Chemical and Drug Induced Liver Injury immunology, Chemical and Drug Induced Liver Injury metabolism, Chemical and Drug Induced Liver Injury pathology, Cytokines metabolism, Disease Models, Animal, Galactosamine, Genetic Predisposition to Disease, Inflammasomes genetics, Inflammasomes immunology, Lipopolysaccharides, Liver drug effects, Liver immunology, Liver pathology, Liver Failure, Acute immunology, Liver Failure, Acute metabolism, Liver Failure, Acute pathology, Macrophage Activation, Macrophages, Peritoneal drug effects, Macrophages, Peritoneal immunology, Macrophages, Peritoneal pathology, Mice, Knockout, NLR Family, Pyrin Domain-Containing 3 Protein genetics, Nuclear Receptor Subfamily 1, Group D, Member 1 agonists, Nuclear Receptor Subfamily 1, Group D, Member 1 deficiency, Nuclear Receptor Subfamily 1, Group D, Member 1 genetics, Peritonitis immunology, Peritonitis metabolism, Peritonitis prevention & control, Phenotype, Pyrrolidines pharmacology, RNA Interference, Severity of Illness Index, Signal Transduction, Thiophenes pharmacology, Time Factors, Transfection, Chemical and Drug Induced Liver Injury prevention & control, Circadian Rhythm, Inflammasomes metabolism, Liver metabolism, Liver Failure, Acute prevention & control, Macrophages, Peritoneal metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Nuclear Receptor Subfamily 1, Group D, Member 1 metabolism
- Abstract
Background & Aims: The innate immune system responds not only to bacterial signals, but also to non-infectious danger-associated molecular patterns that activate the NLRP3 inflammasome complex after tissue injury. Immune functions vary over the course of the day, but it is not clear whether these changes affect the activity of the NLRP3 inflammasome. We investigated whether the core clock component nuclear receptor subfamily 1 group D member 1 (NR1D1, also called Rev-erbα) regulates expression, activity of the NLRP3 inflammasome, and its signaling pathway., Methods: We collected naïve peritoneal macrophages and plasma, at multiple times of day, from Nr1d1
-/- mice and their Nr1d1+/+ littermates (controls) and analyzed expression NLRP3, interleukin 1β (IL1B, in plasma), and IL18 (in plasma). We also collected bone marrow-derived primary macrophages from these mice. Levels of NR1D1 were knocked down with small hairpin RNAs in human primary macrophages. Bone marrow-derived primary macrophages from mice and human primary macrophages were incubated with lipopolysaccharide (LPS) to induce expression of NLRP3, IL1B, and IL18; cells were incubated with LPS and adenosine triphosphate to activate the NLRP3 complex. We analyzed caspase 1 activity and cytokine secretion. NR1D1 was activated in primary mouse and human macrophages by incubation with SR9009; some of the cells were also incubated with an NLRP3 inhibitor or inhibitors of caspase 1. Nr1d1-/- mice and control mice were given intraperitoneal injections of LPS to induce peritoneal inflammation; plasma samples were isolated and levels of cytokines were measured. Nr1d1-/- mice, control mice, and control mice given injections of SR9009 were given LPS and D-galactosamine to induce fulminant hepatitis and MCC950 to specifically inhibit NLRP3; plasma was collected to measure cytokines and a marker of liver failure (alanine aminotransferase); liver tissues were collected and analyzed by quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry., Results: In peritoneal macrophages, expression of NLRP3 and activation of its complex varied with time of day (circadian rhythm)-this regulation required NR1D1. Primary macrophages from Nr1d1-/- mice and human macrophages with knockdown of NR1D1 had altered expression patterns of NLRP3, compared to macrophages that expressed NR1D1, and altered patterns of IL1B and 1L18 production. Mice with disruption of Nr1d1 developed more-severe acute peritoneal inflammation and fulminant hepatitis than control mice. Incubation of macrophage with the NR1D1 activator SR9009 reduced expression of NLRP3 and secretion of cytokines. Mice given SR9009 developed less-severe liver failure and had longer survival times than mice given saline (control)., Conclusions: In studies of Nr1d1-/- mice and human macrophages with pharmacologic activation of NR1D1, we found NR1D1 to regulate the timing of NLRP3 expression and production of inflammatory cytokines by macrophages. Activation of NR1D1 reduced the severity of peritoneal inflammation and fulminant hepatitis in mice., (Copyright © 2018 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
48. REV-ERBβ is required to maintain normal wakefulness and the wake-inducing effect of dual REV-ERB agonist SR9009.
- Author
-
Amador A, Kamenecka TM, Solt LA, and Burris TP
- Subjects
- Animals, Circadian Rhythm drug effects, Electroencephalography drug effects, Electroencephalography methods, Electromyography drug effects, Electromyography methods, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Wakefulness drug effects, Circadian Rhythm physiology, Pyrrolidines pharmacology, Receptors, Cytoplasmic and Nuclear agonists, Receptors, Cytoplasmic and Nuclear deficiency, Repressor Proteins agonists, Repressor Proteins deficiency, Thiophenes pharmacology, Wakefulness physiology
- Abstract
Circadian signaling regulates and synchronizes physiological and behavioral processes, such as feeding, metabolism, and sleep cycles. The endogenous molecular machinery that regulates circadian activities is located in the suprachiasmatic nucleus of the hypothalamus. The REV-ERBs are transcription factors that play key roles in the regulation of the circadian clock and metabolism. Using pharmacological methods, we recently demonstrated the involvement of the REV-ERBs in sleep architecture. Another group reported a delayed response to sleep deprivation and altered sleep cycles in REV-ERBα null mice, indicating a role of REV-ERBα in sleep. Given that REV-ERBβ is structurally and functionally similar to REV-ERBα, we investigated the role of REV-ERBβ in sleep and wakefulness by assessing electroencephalographic recordings in REV-ERBβ deficient mice and the mechanism underlying effects of loss of REV-ERBβ on sleep. Our data suggest that REV-ERBβ is involved in the maintenance of wakefulness during the activity period. In addition, REV-ERBβ-deficient mice administered with dual REV-ERB agonist SR9009, failed to show drug-induced wake increase. Finally, the expression of a number of genes known to mediate sleep and wakefulness were altered in REV-ERBβ null mice., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
49. Adropin: An endocrine link between the biological clock and cholesterol homeostasis.
- Author
-
Ghoshal S, Stevens JR, Billon C, Girardet C, Sitaula S, Leon AS, Rao DC, Skinner JS, Rankinen T, Bouchard C, Nuñez MV, Stanhope KL, Howatt DA, Daugherty A, Zhang J, Schuelke M, Weiss EP, Coffey AR, Bennett BJ, Sethupathy P, Burris TP, Havel PJ, and Butler AA
- Subjects
- Adult, Aged, Animals, Blood Proteins, Cells, Cultured, Female, Glucose metabolism, Hep G2 Cells, Humans, Intercellular Signaling Peptides and Proteins, Liver metabolism, Macaca mulatta, Male, Mice, Middle Aged, Nuclear Receptor Subfamily 1, Group F, Member 1 metabolism, Nuclear Receptor Subfamily 1, Group F, Member 3 metabolism, Proteins genetics, Cholesterol, LDL blood, Circadian Clocks, Homeostasis, Peptides blood, Proteins metabolism
- Abstract
Objective: Identify determinants of plasma adropin concentrations, a secreted peptide translated from the Energy Homeostasis Associated (ENHO) gene linked to metabolic control and vascular function., Methods: Associations between plasma adropin concentrations, demographics (sex, age, BMI) and circulating biomarkers of lipid and glucose metabolism were assessed in plasma obtained after an overnight fast in humans. The regulation of adropin expression was then assessed in silico, in cultured human cells, and in animal models., Results: In humans, plasma adropin concentrations are inversely related to atherogenic LDL-cholesterol (LDL-C) levels in men (n = 349), but not in women (n = 401). Analysis of hepatic Enho expression in male mice suggests control by the biological clock. Expression is rhythmic, peaking during maximal food consumption in the dark correlating with transcriptional activation by RORα/γ. The nadir in the light phase coincides with the rest phase and repression by Rev-erb. Plasma adropin concentrations in nonhuman primates (rhesus monkeys) also exhibit peaks coinciding with feeding times (07:00 h, 15:00 h). The ROR inverse agonists SR1001 and the 7-oxygenated sterols 7-β-hydroxysterol and 7-ketocholesterol, or the Rev-erb agonist SR9009, suppress ENHO expression in cultured human HepG2 cells. Consumption of high-cholesterol diets suppress expression of the adropin transcript in mouse liver. However, adropin over expression does not prevent hypercholesterolemia resulting from a high cholesterol diet and/or LDL receptor mutations., Conclusions: In humans, associations between plasma adropin concentrations and LDL-C suggest a link with hepatic lipid metabolism. Mouse studies suggest that the relationship between adropin and cholesterol metabolism is unidirectional, and predominantly involves suppression of adropin expression by cholesterol and 7-oxygenated sterols. Sensing of fatty acids, cholesterol and oxysterols by the RORα/γ ligand-binding domain suggests a plausible functional link between adropin expression and cellular lipid metabolism. Furthermore, the nuclear receptors RORα/γ and Rev-erb may couple adropin synthesis with circadian rhythms in carbohydrate and lipid metabolism., (Copyright © 2018 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
50. Pharmacological inhibition of REV-ERB stimulates differentiation, inhibits turnover and reduces fibrosis in dystrophic muscle.
- Author
-
Welch RD, Billon C, Valfort AC, Burris TP, and Flaveny CA
- Subjects
- 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, 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
- 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.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.