Network


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

Hotspot


Dive into the research topics where Harold B. Wood is active.

Publication


Featured researches published by Harold B. Wood.


Molecular Pharmacology | 2007

The differential interactions of peroxisome proliferator-activated receptor gamma ligands with Tyr473 is a physical basis for their unique biological activities.

Monica Einstein; Taro E. Akiyama; Gino Castriota; Chuanlin F. Wang; Brian M. McKeever; Ralph T. Mosley; Joseph W. Becker; David E. Moller; Peter T. Meinke; Harold B. Wood; Joel P. Berger

Despite their proven antidiabetic efficacy, widespread use of peroxisome proliferator-activated receptor (PPAR)γ agonists has been limited by adverse cardiovascular effects. To overcome this shortcoming, selective PPARγ modulators (SPPARγMs) have been identified that have antidiabetic efficacy comparable with full agonists with improved tolerability in preclinical species. The results of structural studies support the proposition that SPPARγMs interact with PPARγ differently from full agonists, thereby providing a physical basis for their novel activities. Herein, we describe a novel PPARγ ligand, SPPARγM2. This compound was a partial agonist in a cell-based transcriptional activity assay, with diminished adipogenic activity and an attenuated gene signature in cultured human adipocytes. X-ray cocrystallography studies demonstrated that, unlike rosiglitazone, SPPARγM2 did not interact with the Tyr473 residue located within helix 12 of the ligand binding domain (LBD). Instead, SPPARγM2 was found to bind to and activate human PPARγ in which the Tyr473 residue had been mutated to alanine (hPPARγY473A), with potencies similar to those observed with the wild-type receptor (hPPARγWT). In additional studies, we found that the intrinsic binding and functional potencies of structurally distinct SPPARγMs were not diminished by the Y473A mutation, whereas those of various thiazolidinedione (TZD) and non-TZD PPARγ full agonists were reduced in a correlative manner. These results directly demonstrate the important role of Tyr473 in mediating the interaction of full agonists but not SPPARγMs with the PPARγ LBD, thereby providing a precise molecular determinant for their differing pharmacologies.


Journal of Medicinal Chemistry | 2009

Discovery of (2R)-2-(3-{3-[(4-Methoxyphenyl)carbonyl]-2-methyl-6-(trifluoromethoxy)-1H-indol-1-yl}phenoxy)butanoic Acid (MK-0533): A Novel Selective Peroxisome Proliferator-Activated Receptor γ Modulator for the Treatment of Type 2 Diabetes Mellitus with a Reduced Potential to Increase Plasma and Extracellular Fluid Volume

John J. Acton; Taro E. Akiyama; Ching H. Chang; Lawrence F. Colwell; Sheryl D. Debenham; Thomas W. Doebber; Monica Einstein; Kun Liu; Margaret E. McCann; David E. Moller; Eric S. Muise; Yejun Tan; John R. Thompson; Kenny K. Wong; Margaret Wu; Libo Xu; Peter T. Meinke; Joel P. Berger; Harold B. Wood

Peroxisome proliferator-activated receptor gamma (PPARgamma) agonists are used to treat type 2 diabetes mellitus (T2DM). Widespread use of PPARgamma agonists has been prevented due to adverse effects including weight gain, edema, and increased risk of congestive heart failure. Selective PPARgamma modulators (SPPARgammaMs) have been identified that have antidiabetic efficacy and reduced toxicity in preclinical species. In comparison with PPARgamma full agonists, SPPARgammaM 6 (MK0533) displayed diminished maximal activity (partial agonism) in cell-based transcription activation assays and attenuated gene signatures in adipose tissue. Compound 6 exhibited comparable efficacy to rosiglitazone and pioglitazone in vivo. However, with regard to the induction of untoward events, 6 displayed no cardiac hypertrophy, attenuated increases in brown adipose tissue, minimal increases in plasma volume, and no increases in extracellular fluid volume in vivo. Further investigation of 6 is warranted to determine if the improvement in mechanism-based side effects observed in preclinical species will be recapitulated in humans.


Bioorganic & Medicinal Chemistry Letters | 2011

Design of potent and selective GPR119 agonists for type II diabetes.

Jason W. Szewczyk; John J. Acton; Alan D. Adams; Gary G. Chicchi; Stanley Freeman; Andrew D. Howard; Yong Huang; Cai Li; Peter T. Meinke; Ralph Mosely; Elizabeth Murphy; Rachel Samuel; Conrad Santini; Meng Yang; Yong Zhang; Kake Zhao; Harold B. Wood

Screening of the Merck sample collection identified compound 1 as a weakly potent GPR119 agonist (hEC(50)=3600 nM). Dual termini optimization of 1 led to compound 36 having improved potency, selectivity, and formulation profile, however, modest physical properties (PP) hindered its utility. Design of a new core containing a cyclopropyl restriction yielded further PP improvements and when combined with the termini SAR optimizations yielded a potent and highly selective agonist suitable for further preclinical development (58).


Journal of Medicinal Chemistry | 2011

Benzimidazolones: A New Class of Selective Peroxisome Proliferator-Activated Receptor γ (PPARγ) Modulators

Weiguo Liu; Fiona WaiYu Lau; Kun Liu; Harold B. Wood; Gaochao Zhou; Yuli Chen; Ying Li; Taro E. Akiyama; Gino Castriota; Monica Einstein; Chualin Wang; Margaret E. McCann; Thomas W. Doebber; Margaret Wu; Ching H. Chang; Brian M. McKeever; Ralph T. Mosley; Joel P. Berger; Peter T. Meinke

A series of benzimidazolone carboxylic acids and oxazolidinediones were designed and synthesized in search of selective PPARγ modulators (SPPARγMs) as potential therapeutic agents for the treatment of type II diabetes mellitus (T2DM) with improved safety profiles relative to rosiglitazone and pioglitazone, the currently marketed PPARγ full agonist drugs. Structure-activity relationships of these potent and highly selective SPPARγMs were studied with a focus on their unique profiles as partial agonists or modulators. A variety of methods, such as X-ray crystallographic analysis, PPARγ transactivation coactivator profiling, gene expression profiling, and mutagenesis studies, were employed to reveal the differential interactions of these new analogues with PPARγ receptor in comparison to full agonists. In rodent models of T2DM, benzimidazolone analogues such as (5R)-5-(3-{[3-(5-methoxybenzisoxazol-3-yl)benzimidazol-1-yl]methyl}phenyl)-5-methyloxazolidinedione (51) demonstrated efficacy equivalent to that of rosiglitazone. Side effects, such as fluid retention and heart weight gain associated with PPARγ full agonists, were diminished with 51 in comparison to rosiglitazone based on studies in two independent animal models.


European Journal of Pharmacology | 2008

A novel selective peroxisome proliferator-activator receptor-γ modulator—SPPARγM5 improves insulin sensitivity with diminished adverse cardiovascular effects

Ching H. Chang; Margaret Wu; Eric S. Muise; Yejun Tan; Harold B. Wood; Peter T. Meinke; John R. Thompson; Tom Doebber; Joel P. Berger; Margaret E. McCann

The use of the thiazolidinedione insulin sensitizers rosiglitazone and pioglitazone for the treatment of type 2 diabetes mellitus in recent years has proven to be effective in helping patients resume normal glycemic control. However, their use is often associated with undesirable side effects including peripheral edema, congestive heart failure and weight gain. Here, we report the identification and characterization of a novel selective PPARgamma modulator, SPPARgammaM5 ((2S)-2-(2-chloro-5-{[3-(4-chlorophenoxy)-2-methyl-6-(trifluoromethoxy)-1H-indol-1-yl]methyl} phenoxy)propionic acid), which has notable insulin sensitizing properties and a superior tolerability profile to that of rosiglitazone. SPPARgammaM5 is a potent ligand of human PPARgamma with high selectivity versus PPARalpha or PPARdelta in receptor competitive binding assays. In cell-based transcriptional activation assays, SPPARgammaM5 was a potent partial agonist of human PPARgamma in comparison to the PPARgamma full agonist rosiglitazone. Compared to rosiglitazone or the PPARgamma full agonist COOH (2-(2-(4-phenoxy-2-propylphenoxy)ethyl)indole-5-acetic acid), SPPARgammaM5 induced an attenuated PPARgamma-regulated gene expression profile in fully differentiated 3T3-L1 adipocytes and white adipose tissue of chronically treated db/db mice. SPPARgammaM5 treatment also reduced the insulin resistance index by homeostasis model assessment (HOMA), suggesting an improvement in insulin resistance in these db/db mice. Treatment of obese Zucker rats with either rosiglitazone or SPPARgammaM5 resulted in an improvement in selected parameters that serve as surrogate indicators of insulin resistance and hyperlipidemia. However, unlike rosiglitazone, SPPARgammaM5 did not cause significant fluid retention or cardiac hypertrophy in these rats. Thus, compounds such as SPPARgammaM5 may offer beneficial effects on glycemic control with significantly attenuated adverse effects.


Journal of Medicinal Chemistry | 2009

Activation of Peroxisome Proliferator-Activated Receptor γ (PPARγ) by Nitroalkene Fatty Acids: Importance of Nitration Position and Degree of Unsaturation

Michael J. Gorczynski; Pamela K. Smitherman; Taro E. Akiyama; Harold B. Wood; Joel P. Berger; S. Bruce King; Charles S. Morrow

Nitroalkene fatty acids are potent endogenous ligand activators of PPARgamma-dependent transcription. Previous studies with the naturally occurring regioisomers of nitrolinoleic acid revealed that the isomers are not equivalent with respect to PPARgamma activation. To gain further insight into the structure-activity relationships between nitroalkenes and PPARgamma, we examined additional naturally occurring nitroalkenes derived from oleic acid, 9-nitrooleic acid (E-9-NO2-18:1 [1]) and 10-nitrooleic acid (E-10-NO2-18:1 [2]), and several synthetic nitrated enoic fatty acids of variable carbon chain length, double bonds, and nitration site. At submicromolar concentrations, E-12-NO2 derivatives were considerably more potent than isomers nitrated at carbons 5, 6, 9, 10, and 13, and chain length (16 versus 18) or number of double bonds (1 versus 2) was of little consequence for PPARgamma activation. Interestingly, at higher concentrations (>2 microM) the nitrated enoic fatty acids (E-9-NO2-18:1 [1], E-9-NO2-16:1 [3], E-10-NO2-18:1 [2], and E-12-NO2-18:1 [7]) deviated significantly from the saturable pattern of PPARgamma activation observed for nitrated 1,4-dienoic fatty acids (E-9-NO2-18:2, E-10-NO2-18:2, E-12-NO2-18:2, and E-13-NO2-18:2).


Tetrahedron Letters | 1999

Total synthesis of asterriquinone B1

Kun Liu; Harold B. Wood; A. Brian Jones

Abstract This communication describes the first total synthesis of asterriquinone B1, a representative member of a group of anti-tumor metabolites of Aspergillus terreus . The synthesis described herein is potentially applicable to other members of the asterriquinone family.


Journal of Medicinal Chemistry | 2009

Discovery of a Peroxisome Proliferator Activated Receptor γ (PPARγ) Modulator with Balanced PPARα Activity for the Treatment of Type 2 Diabetes and Dyslipidemia

Weiguo Liu; Kun Liu; Harold B. Wood; Margaret E. McCann; Thomas W. Doebber; Ching H. Chang; Taro E. Akiyama; Monica Einstein; Joel P. Berger; Peter T. Meinke

A series of 3-acylindole-1-benzylcarboxylic acids were designed and synthesized while searching for a PPARgamma modulator with additional moderate intrinsic PPARalpha agonistic activity. 2-[3-[[3-(4-Chlorobenzoyl)-2-methyl-6-(trifluoromethoxy)-1H-indol-1-yl]methyl]phenoxy]-(2R)-butanoic acid (12d) was identified as such an agent which demonstrated potent efficacy in lowering both glucose and lipids in multiple animal models with significantly attenuated side effects such as fluid retention and heart weight gain associated with PPARgamma full agonists. The moderate PPARalpha activity of 12d not only contributed to the agents ability to manage lipid profiles but also appears to have potentiated its PPARgamma efficacy in lowering glucose levels in preclinical diabetic animal models.


Bioorganic & Medicinal Chemistry Letters | 2000

The basal SAR of a novel insulin receptor activator.

Harold B. Wood; Regina M. Black; Gino Salituro; Deborah Szalkowski; Zhihua Li; Yan Zhang; David E. Moller; Bei Zhang; A. Brian Jones

The synthesis and SAR of analogues prepared from novel insulin receptor activator 1 are described. Changes to the dihydroxyquinone core were not tolerated while functionalization of the two indoles contained in 1 resulted in little effect upon activation of the insulin receptor.


Annual Reports in Medicinal Chemistry | 2006

Nuclear Hormone Receptor Modulators for the Treatment of Diabetes and Dyslipidemia

Peter T. Meinke; Harold B. Wood; Jason W. Szewczyk

Publisher Summary Concerted endeavors to identify superior methods to treat multiple aspects of the metabolic syndrome have fueled a veritable renaissance in the field of nuclear hormone receptor (NHR) modulators. These biological targets, which include the peroxisome-proliferator-activated receptors (PPAR), retinoid X receptor (RXR), liver X receptor (LXR), and farnesoid X receptor (FXR), are recognized as pharmaceutically viable intervention points. The PPAR subtype family comprises three members (α, γ, and δ) that function as lipid sensors and transcriptional regulators of nutrient homeostasis. RXR heterodimers with PPAR, LXR, and FXR are known to activate gene transcription in the presence of an agonist for either of the partnered receptors. The NHR LXR comprises two subtypes—LXRa (NR1H3) and LXRb (NR1H2)—acting as intracellular oxysterol sensors that regulate the expression of genes involved in lipid metabolism. FXR is an NHR acting as a physiological sensor for bile acids by controlling genes regulating their metabolism and transport. The repression of key genes, Cyp7A1 and Cyp8B1, by FXR holds much of its promise as a therapeutic target.

Researchain Logo
Decentralizing Knowledge