Network


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

Hotspot


Dive into the research topics where Clarence Hale is active.

Publication


Featured researches published by Clarence Hale.


Diabetes | 2009

Fibroblast Growth Factor 21 Reverses Hepatic Steatosis, Increases Energy Expenditure, and Improves Insulin Sensitivity in Diet-Induced Obese Mice

Jing Xu; David J. Lloyd; Clarence Hale; Shanaka Stanislaus; Michelle Chen; Glenn Sivits; Steven Vonderfecht; Randy Ira Hecht; Yue-Sheng Li; Richard Lindberg; Jin-Long Chen; Dae Young Jung; Zhiyou Zhang; Hwi Jin Ko; Jason K. Kim; Murielle M. Véniant

OBJECTIVE—Fibroblast growth factor 21 (FGF21) has emerged as an important metabolic regulator of glucose and lipid metabolism. The aims of the current study are to evaluate the role of FGF21 in energy metabolism and to provide mechanistic insights into its glucose and lipid-lowering effects in a high-fat diet–induced obesity (DIO) model. RESEARCH DESIGN AND METHODS—DIO or normal lean mice were treated with vehicle or recombinant murine FGF21. Metabolic parameters including body weight, glucose, and lipid levels were monitored, and hepatic gene expression was analyzed. Energy metabolism and insulin sensitivity were assessed using indirect calorimetry and hyperinsulinemic-euglycemic clamp techniques. RESULTS—FGF21 dose dependently reduced body weight and whole-body fat mass in DIO mice due to marked increases in total energy expenditure and physical activity levels. FGF21 also reduced blood glucose, insulin, and lipid levels and reversed hepatic steatosis. The profound reduction of hepatic triglyceride levels was associated with FGF21 inhibition of nuclear sterol regulatory element binding protein-1 and the expression of a wide array of genes involved in fatty acid and triglyceride synthesis. FGF21 also dramatically improved hepatic and peripheral insulin sensitivity in both lean and DIO mice independently of reduction in body weight and adiposity. CONCLUSIONS—FGF21 corrects multiple metabolic disorders in DIO mice and has the potential to become a powerful therapeutic to treat hepatic steatosis, obesity, and type 2 diabetes.


Endocrinology | 2012

Lack of Overt FGF21 Resistance in Two Mouse Models of Obesity and Insulin Resistance

Clarence Hale; Michelle Chen; Shanaka Stanislaus; Narumol Chinookoswong; Todd Hager; Minghan Wang; Murielle M. Véniant; Jing Xu

Circulating levels of fibroblast growth factor 21 (FGF21), a metabolic regulator of glucose, lipid, and energy homeostasis, are elevated in obese diabetic subjects, raising questions about potential FGF21 resistance. Here we report tissue expression changes in FGF21 and its receptor components, and we describe the target-organ and whole-body responses to FGF21 in ob/ob and diet-induced obese (DIO) mice. Plasma FGF21 concentrations were elevated 8- and 16-fold in DIO and ob/ob mice, respectively, paralleling a dramatic increase in hepatic FGF21 mRNA expression. Concurrently, expression levels of βKlotho, FGF receptor (FGFR)-1c, and FGFR2c were markedly down-regulated in the white adipose tissues (WAT) of ob/ob and DIO mice. However, dose-response curves of recombinant human FGF21 (rhFGF21) stimulation of ERK phosphorylation in the liver and WAT were not right shifted in disease models, although the magnitude of induction in ERK phosphorylation was partially attenuated in DIO mice. Whole-body metabolic responses were preserved in ob/ob and DIO mice, with disease models being more sensitive and responsive than lean mice to the glucose-lowering and weight-loss effects of rhFGF21. Endogenous FGF21 levels, although elevated in diseased mice, were below the half-maximal effective concentrations of rhFGF21, suggesting a state of relative deficiency. Hepatic and WAT FGF21 mRNA expression levels declined after rhFGF21 treatment in the absence of the increased expression levels of βKlotho and FGFR. We conclude that overt FGF21 resistance was not evident in the disease models, and increased hepatic FGF21 expression as a result of local metabolic changes is likely a major cause of elevated circulating FGF21 levels.


PLOS ONE | 2012

FGF21 Promotes Metabolic Homeostasis via White Adipose and Leptin in Mice

Murielle M. Véniant; Clarence Hale; Joan Helmering; Michelle Chen; Shanaka Stanislaus; Jim Busby; Steven Vonderfecht; Jing Xu; David J. Lloyd

Fibroblast growth factor 21 (FGF21) is a potent metabolic regulator, and pharmacological administration elicits glucose and lipid lowering responses in mammals. To delineate if adipose tissue is the predominant organ responsible for anti-diabetic effects of FGF21, we treated mice with reduced body fat (lipodystrophy mice with adipose specific expression of active sterol regulatory element binding protein 1c; Tg) with recombinant murine FGF21 (rmuFGF21). Unlike wildtype (WT) mice, Tg mice were refractory to the beneficial effects of rmuFGF21 on body weight, adipose mass, plasma insulin and glucose tolerance. To determine if adipose mass was critical for these effects, we transplanted WT white adipose tissue (WAT) into Tg mice and treated the mice with rmuFGF21. After transplantation, FGF21 responsiveness was completely restored in WAT transplanted Tg mice compared to sham Tg mice. Further, leptin treatment alone was sufficient to restore the anti-diabetic effects of rmuFGF21 in Tg mice. Molecular analyses of Tg mice revealed normal adipose expression of Fgfr1, Klb and an 8-fold over-expression of Fgf21. Impaired FGF21-induced signaling indicated that residual adipose tissue of Tg mice was resistant to FGF21, whilst normal FGF21 signaling was observed in Tg livers. Together these data suggest that adipose tissue is required for the triglyceride and glucose, but not the cholesterol lowering efficacy of FGF21, and that leptin and FGF21 exert additive anti-diabetic effects in Tg mice.


Nature | 2013

Antidiabetic effects of glucokinase regulatory protein small-molecule disruptors

David J. Lloyd; David J. St. Jean; Robert J.M. Kurzeja; Robert C. Wahl; Klaus Michelsen; Rod Cupples; Michelle Chen; John Wu; Glenn Sivits; Joan Helmering; Renee Komorowski; Kate S. Ashton; Lewis D. Pennington; Christopher Fotsch; Mukta Vazir; Kui Chen; Samer Chmait; Jiandong Zhang; Longbin Liu; Mark H. Norman; Kristin L. Andrews; Michael D. Bartberger; Gwyneth Van; Elizabeth J. Galbreath; Steven Vonderfecht; Minghan Wang; Steven R. Jordan; Murielle M. Véniant; Clarence Hale

Glucose homeostasis is a vital and complex process, and its disruption can cause hyperglycaemia and type II diabetes mellitus. Glucokinase (GK), a key enzyme that regulates glucose homeostasis, converts glucose to glucose-6-phosphate in pancreatic β-cells, liver hepatocytes, specific hypothalamic neurons, and gut enterocytes. In hepatocytes, GK regulates glucose uptake and glycogen synthesis, suppresses glucose production, and is subject to the endogenous inhibitor GK regulatory protein (GKRP). During fasting, GKRP binds, inactivates and sequesters GK in the nucleus, which removes GK from the gluconeogenic process and prevents a futile cycle of glucose phosphorylation. Compounds that directly hyperactivate GK (GK activators) lower blood glucose levels and are being evaluated clinically as potential therapeutics for the treatment of type II diabetes mellitus. However, initial reports indicate that an increased risk of hypoglycaemia is associated with some GK activators. To mitigate the risk of hypoglycaemia, we sought to increase GK activity by blocking GKRP. Here we describe the identification of two potent small-molecule GK–GKRP disruptors (AMG-1694 and AMG-3969) that normalized blood glucose levels in several rodent models of diabetes. These compounds potently reversed the inhibitory effect of GKRP on GK activity and promoted GK translocation both in vitro (isolated hepatocytes) and in vivo (liver). A co-crystal structure of full-length human GKRP in complex with AMG-1694 revealed a previously unknown binding pocket in GKRP distinct from that of the phosphofructose-binding site. Furthermore, with AMG-1694 and AMG-3969 (but not GK activators), blood glucose lowering was restricted to diabetic and not normoglycaemic animals. These findings exploit a new cellular mechanism for lowering blood glucose levels with reduced potential for hypoglycaemic risk in patients with type II diabetes mellitus.


Journal of Medicinal Chemistry | 2010

Discovery of a Potent, Orally Active 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor for Clinical Study: Identification of (S)-2-((1S,2S,4R)-Bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221)

Murielle M. Véniant; Clarence Hale; Randall W. Hungate; Kyung Gahm; Maurice Emery; Janan Jona; Smriti Joseph; Jeffrey Adams; Andrew Hague; George A. Moniz; Jiandong Zhang; Michael D. Bartberger; Vivian Li; Rashid Syed; Steven R. Jordan; Renee Komorowski; Michelle Chen; Rod Cupples; Ki Won Kim; David J. St. Jean; Lars Johansson; Martin Henriksson; Meredith Williams; Jerk Vallgarda; Christopher Fotsch; Minghan Wang

Thiazolones with an exo-norbornylamine at the 2-position and an isopropyl group on the 5-position are potent 11beta-HSD1 inhibitors. However, the C-5 center was prone to epimerization in vitro and in vivo, forming a less potent diastereomer. A methyl group was added to the C-5 position to eliminate epimerization, leading to the discovery of (S)-2-((1S,2S,4R)-bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221). This compound decreased fed blood glucose and insulin levels and reduced body weight in diet-induced obesity mice.


Diabetes, Obesity and Metabolism | 2009

Antidiabetic effects of 11β-HSD1 inhibition in a mouse model of combined diabetes, dyslipidaemia and atherosclerosis

David J. Lloyd; Joan Helmering; D. Cordover; M. Bowsman; Michelle Chen; Clarence Hale; P. Fordstrom; M. Zhou; Minghan Wang; Stephen Kaufman; Murielle M. Véniant

Aim:  11 β‐hydroxysteroid dehydrogenase type 1 (11β‐HSD1) is considered to contribute to the aetiology of the metabolic syndrome, and specific inhibitors have begun to emerge as treatments for insulin resistance and other facets of the syndrome, including atherosclerosis. Given the role of glucocorticoids and 11β‐HSD1 in the anti‐inflammatory response and the involvement of inflammation in the development of atherosclerosis, 11β‐HSD1 inhibition may exacerbate atherosclerosis. Our aim was to investigate in vivo the effects of a specific 11β‐HSD1 inhibitor (2922) on atherosclerosis while assessing glucose homeostasis.


Journal of Medicinal Chemistry | 2008

2-amino-1,3-thiazol-4(5H)-ones as potent and selective 11beta-hydroxysteroid dehydrogenase type 1 inhibitors: enzyme-ligand co-crystal structure and demonstration of pharmacodynamic effects in C57Bl/6 mice.

Lars Johansson; Christopher Fotsch; Michael D. Bartberger; Victor M. Castro; Michelle Chen; Maurice Emery; Sonja Gustafsson; Clarence Hale; Dean Hickman; Evert Homan; Steven R. Jordan; Renee Komorowski; Aiwen Li; Kenneth McRae; George A. Moniz; Guy Matsumoto; Carlos Orihuela; Gunnar Palm; Murielle M. Véniant; Minghan Wang; Meredith Williams; Jiandong Zhang

11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) has attracted considerable attention during the past few years as a potential target for the treatment of diseases associated with metabolic syndrome. In our ongoing work on 11beta-HSD1 inhibitors, a series of new 2-amino-1,3-thiazol-4(5 H)-ones were explored. By inserting various cycloalkylamines at the 2-position and alkyl groups or spirocycloalkyl groups at the 5-position of the thiazolone, several potent 11beta-HSD1 inhibitors were identified. An X-ray cocrystal structure of human 11beta-HSD1 with compound 6d (Ki=28 nM) revealed a large lipophilic pocket accessible by substitution off the 2-position of the thiazolone. To increase potency, analogues were prepared with larger lipophilic groups at this position. One of these compounds, the 3-noradamantyl analogue 8b, was a potent inhibitor of human 11beta-HSD1 (Ki=3 nM) and also inhibited 11beta-HSD1 activity in lean C57Bl/6 mice when evaluated in an ex vivo adipose and liver cortisone to cortisol conversion assay.


Bioorganic & Medicinal Chemistry Letters | 2003

Design of a new peptidomimetic agonist for the melanocortin receptors based on the solution structure of the peptide ligand, Ac-Nle-cyclo[Asp-Pro-dPhe-Arg-Trp-Lys]-NH2

Christopher Fotsch; Duncan M. Smith; Jeffrey Adams; Janet Cheetham; Michael Croghan; Elizabeth M. Doherty; Clarence Hale; Mark A. Jarosinski; Michael G. Kelly; Mark H. Norman; Nuria A. Tamayo; Ning Xi; James W. Baumgartner

The solution structure of a potent melanocortin receptor agonist, Ac-Nle-cyclo[Asp-Pro-DPhe-Arg-Trp-Lys]-NH(2) (1) was calculated using distance restraints determined from 1H NMR spectroscopy. Eight of the lowest energy conformations from this study were used to identify non-peptide cores that mimic the spatial arrangement of the critical tripeptide region, DPhe-Arg-Trp, found in 1. From these studies, compound 2a, containing the cis-cyclohexyl core, was identified as a functional agonist of the melanocortin-4 receptor (MC4R) with an IC(50) and EC(50) below 10 nM. Compound 2a also showed 36- and 7-fold selectivity over MC3R and MC1R, respectively, in the binding assays. Subtle changes in cyclohexane stereochemistry and removal of functional groups led to analogues with lower affinity for the MC receptors.


Diabetes, Obesity and Metabolism | 2009

Time of the day for 11β‐HSD1 inhibition plays a role in improving glucose homeostasis in DIO mice

Murielle M. Véniant; Clarence Hale; Renee Komorowski; Michelle Chen; D. J. St Jean; Christopher Fotsch; Minghan Wang

Aims:  The physiological effects of glucocorticoids in a given tissue are driven by the local level of the active glucocorticoid, which is determined by two sources: the plasma cortisol in human (or corticosterone in rodents) and the cortisol produced locally through 11β‐hydroxysteroid dehydrogenase type 1 (11β‐HSD1) activity. Because of the circadian variation of plasma glucocorticoids, the pharmacological efficacy of 11β‐HSD1 inhibition may depend on the time of the day for inhibitor administration.


Chemical Biology & Drug Design | 2007

Structural characterization and pharmacodynamic effects of an orally active 11beta-hydroxysteroid dehydrogenase type 1 inhibitor.

Clarence Hale; Murielle M. Véniant; Zhulun Wang; Michelle Chen; Jocelyn McCormick; Rod Cupples; Dean Hickman; Xiaoshan Min; Athena Sudom; Haoda Xu; Guy Matsumoto; Christopher Fotsch; David J. St. Jean; Minghan Wang

11β‐Hydroxysteroid dehydrogenase type 1 regulates glucocorticoid action and inhibition of this enzyme is a viable therapeutic strategy for the treatment of type 2 diabetes and the metabolic syndrome. Here, we report a potent and selective 11β‐hydroxysteroid dehydrogenase type 1 inhibitor with a binding mode elucidated from the co‐crystal structure with the human 11β‐hydroxysteroid dehydrogenase type 1. The inhibitor is bound to the steroid‐binding pocket making contacts with the catalytic center and the solvent channel. The inhibitor binding is facilitated by two direct hydrogen bond interactions involving Tyrosine183 of the catalytic motif Tyr‐X‐X‐X‐Lys and Alanine172. In addition, the inhibitor makes many hydrophobic interactions with both the enzyme and the co‐factor nicotinamide adenine dinucleotide phosphate (reduced). In lean C57BL/6 mice, the compound inhibited both the in vivo and ex vivo 11β‐hydroxysteroid dehydrogenase type 1 activities in a dose‐dependent manner. The inhibitory effects correlate with the plasma compound concentrations, suggesting that there is a clear pharmacokinetic and pharmacodynamic relationship. Moreover, at the same doses used in the pharmacokinetic/pharmacodynamic studies, the inhibitor did not cause the activation of the hypothalamic–pituitary–adrenal axis in an acute mouse model, suggesting that this compound exhibits biological effects with minimal risk of activating the hypothalamic–pituitary–adrenal axis.

Collaboration


Dive into the Clarence Hale's collaboration.

Researchain Logo
Decentralizing Knowledge