Network


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

Hotspot


Dive into the research topics where Aleksandr Petrov is active.

Publication


Featured researches published by Aleksandr Petrov.


Diabetes | 2009

Glucagon-Like Peptide 1/Glucagon Receptor Dual Agonism Reverses Obesity in Mice

Alessandro Pocai; Paul E. Carrington; Jennifer R. Adams; Michael Wright; George J. Eiermann; Lan Zhu; Xiaobing Du; Aleksandr Petrov; Guoqiang Jiang; Franklin Liu; Corey Miller; Laurie Tota; Gaochao Zhou; Xiaoping Zhang; Michael M. Sountis; Alessia Santoprete; Elena Capito; Gary G. Chicchi; Nancy A. Thornberry; Elisabetta Bianchi; Antonello Pessi; Donald J. Marsh; Ranabir SinhaRoy

OBJECTIVE Oxyntomodulin (OXM) is a glucagon-like peptide 1 (GLP-1) receptor (GLP1R)/glucagon receptor (GCGR) dual agonist peptide that reduces body weight in obese subjects through increased energy expenditure and decreased energy intake. The metabolic effects of OXM have been attributed primarily to GLP1R agonism. We examined whether a long acting GLP1R/GCGR dual agonist peptide exerts metabolic effects in diet-induced obese mice that are distinct from those obtained with a GLP1R-selective agonist. RESEARCH DESIGN AND METHODS We developed a protease-resistant dual GLP1R/GCGR agonist, DualAG, and a corresponding GLP1R-selective agonist, GLPAG, matched for GLP1R agonist potency and pharmacokinetics. The metabolic effects of these two peptides with respect to weight loss, caloric reduction, glucose control, and lipid lowering, were compared upon chronic dosing in diet-induced obese (DIO) mice. Acute studies in DIO mice revealed metabolic pathways that were modulated independent of weight loss. Studies in Glp1r−/− and Gcgr−/− mice enabled delineation of the contribution of GLP1R versus GCGR activation to the pharmacology of DualAG. RESULTS Peptide DualAG exhibits superior weight loss, lipid-lowering activity, and antihyperglycemic efficacy comparable to GLPAG. Improvements in plasma metabolic parameters including insulin, leptin, and adiponectin were more pronounced upon chronic treatment with DualAG than with GLPAG. Dual receptor agonism also increased fatty acid oxidation and reduced hepatic steatosis in DIO mice. The antiobesity effects of DualAG require activation of both GLP1R and GCGR. CONCLUSIONS Sustained GLP1R/GCGR dual agonism reverses obesity in DIO mice and is a novel therapeutic approach to the treatment of obesity.


Diabetes | 2008

Selective Small-Molecule Agonists of G Protein–Coupled Receptor 40 Promote Glucose-Dependent Insulin Secretion and Reduce Blood Glucose in Mice

Carina P. Tan; Yue Feng; Yun-Ping Zhou; George J. Eiermann; Aleksandr Petrov; Changyou Zhou; Songnian Lin; Gino Salituro; Peter T. Meinke; Ralph T. Mosley; Taro E. Akiyama; Monica Einstein; Sanjeev Kumar; Joel P. Berger; Sander G. Mills; Nancy A. Thornberry; Lihu Yang; Andrew D. Howard

OBJECTIVE— Acute activation of G protein–coupled receptor 40 (GPR40) by free fatty acids (FFAs) or synthetic GPR40 agonists enhances insulin secretion. However, it is still a matter of debate whether activation of GPR40 would be beneficial for the treatment of type 2 diabetes, since chronic exposure to FFAs impairs islet function. We sought to evaluate the specific role of GPR40 in islets and its potential as a therapeutic target using compounds that specifically activate GPR40. RESEARCH DESIGN AND METHODS— We developed a series of GPR40-selective small-molecule agonists and studied their acute and chronic effects on glucose-dependent insulin secretion (GDIS) in isolated islets, as well as effects on blood glucose levels during intraperitoneal glucose tolerance tests in wild-type and GPR40 knockout mice (GPR40−/−). RESULTS— Small-molecule GPR40 agonists significantly enhanced GDIS in isolated islets and improved glucose tolerance in wild-type mice but not in GPR40−/− mice. While a 72-h exposure to FFAs in tissue culture significantly impaired GDIS in islets from both wild-type and GPR40−/− mice, similar exposure to the GPR40 agonist did not impair GDIS in islets from wild-type mice. Furthermore, the GPR40 agonist enhanced insulin secretion in perfused pancreata from neonatal streptozotocin-induced diabetic rats and improved glucose levels in mice with high-fat diet–induced obesity acutely and chronically. CONCLUSIONS— GPR40 does not mediate the chronic toxic effects of FFAs on islet function. Pharmacological activation of GPR40 may potentiate GDIS in humans and be beneficial for overall glucose control in patients with type 2 diabetes.


European Journal of Pharmacology | 2009

Inhibition of DPP-4 with sitagliptin improves glycemic control and restores islet cell mass and function in a rodent model of type 2 diabetes.

James Mu; Aleksandr Petrov; George J. Eiermann; John Woods; Yun-Ping Zhou; Zhihua Li; Emanuel Zycband; Yue Feng; Lan Zhu; Ranabir Sinha Roy; Andrew D. Howard; Cai Li; Nancy A. Thornberry; Bei B. Zhang

Inhibition of dipeptidyl peptidase-4 (DPP-4) activity has been shown to improve glycemic control in patients with type 2 diabetes by prolonging and potentiating the actions of incretin hormones. This study is designed to determine the effects of the DPP-4 inhibitor sitagliptin on improving islet function in a mouse model of insulin resistance and insulin secretion defects. ICR mice were pre-treated with high fat diet and a low dose of streptozotocin to induce insulin resistance and impaired insulin secretion, respectively. Diabetic mice were treated with sitagliptin or the sulfonylurea agent glipizide as admixture to high fat diet for ten weeks. Sustained reduction of blood glucose, HbA(1c), circulating glucagon and improvement in oral glucose tolerance were observed in mice treated with sitagliptin. In contrast, glipizide improved glycemic control only during the early weeks and to a lesser degree compared to sitagliptin, and had no effect on circulating glucagon levels or glucose tolerance. The improvement in glycemic control in sitagliptin-treated mice was associated with a significant increase in glucose-dependent insulin secretion in both perfused pancreas and isolated islets. Importantly, in contrast to the lack of effect by glipizide, sitagliptin significantly restored beta and alpha cell mass as well as alpha/beta cell ratio. These data indicate that DPP-4 inhibition by sitagliptin provided better overall improvement of glycemic control compared to glipizide in the high fat diet/streptozotocin induced diabetic mouse model. The ability of sitagliptin to enhance islet cell function may offer insight into the potential for disease modification.


Journal of Medicinal Chemistry | 2014

Omarigliptin (MK-3102): A Novel Long-Acting DPP-4 Inhibitor for Once-Weekly Treatment of Type 2 Diabetes.

Tesfaye Biftu; Ranabir SinhaRoy; Ping Chen; Xiaoxia Qian; Dennis Feng; Jeffrey T. Kuethe; Giovanna Scapin; Ying Duo Gao; Youwei Yan; Davida Krueger; Annette Bak; George J. Eiermann; Jiafang He; Jason M. Cox; Jacqueline D. Hicks; Kathy Lyons; Huaibing He; Gino Salituro; Sharon Tong; Sangita B. Patel; George A. Doss; Aleksandr Petrov; Joe C. Wu; Shiyao Sherrie Xu; Charles Sewall; Xiaoping Zhang; Bei Zhang; Nancy A. Thornberry; Ann E. Weber

In our effort to discover DPP-4 inhibitors with added benefits over currently commercially available DPP-4 inhibitors, MK-3102 (omarigliptin), was identified as a potent and selective dipeptidyl peptidase 4 (DPP-4) inhibitor with an excellent pharmacokinetic profile amenable for once-weekly human dosing and selected as a clinical development candidate. This manuscript summarizes the mechanism of action, scientific rationale, medicinal chemistry, pharmacokinetic properties, and human efficacy data for omarigliptin, which is currently in phase 3 clinical development.


Journal of Lipid Research | 2012

Plasma lipid profiling across species for the identification of optimal animal models of human dyslipidemia.

Wu Yin; Ester Carballo-Jane; David G. McLaren; Vivienne Mendoza; Karen Gagen; Neil S. Geoghagen; Judith N. Gorski; George J. Eiermann; Aleksandr Petrov; Michael Wolff; Xinchun Tong; Larissa Wilsie; Taro E. Akiyama; Jing Chen; Anil Thankappan; Jiyan Xue; Xiaoli Ping; Genevieve Andrews; L. Alexandra Wickham; Cesaire L. Gai; Tu Trinh; Alison Kulick; Marcie J. Donnelly; Gregory O. Voronin; Ray Rosa; Anne-Marie Cumiskey; Kavitha Bekkari; Lyndon J. Mitnaul; Oscar Puig; Fabian Chen

In an attempt to understand the applicability of various animal models to dyslipidemia in humans and to identify improved preclinical models for target discovery and validation for dyslipidemia, we measured comprehensive plasma lipid profiles in 24 models. These included five mouse strains, six other nonprimate species, and four nonhuman primate (NHP) species, and both healthy animals and animals with metabolic disorders. Dyslipidemic humans were assessed by the same measures. Plasma lipoprotein profiles, eight major plasma lipid fractions, and FA compositions within these lipid fractions were compared both qualitatively and quantitatively across the species. Given the importance of statins in decreasing plasma low-density lipoprotein cholesterol for treatment of dyslipidemia in humans, the responses of these measures to simvastatin treatment were also assessed for each species and compared with dyslipidemic humans. NHPs, followed by dog, were the models that demonstrated closest overall match to dyslipidemic humans. For the subset of the dyslipidemic population with high plasma triglyceride levels, the data also pointed to hamster and db/db mouse as representative models for practical use in target validation. Most traditional models, including rabbit, Zucker diabetic fatty rat, and the majority of mouse models, did not demonstrate overall similarity to dyslipidemic humans in this study.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of 5-aryloxy-2,4-thiazolidinediones as potent GPR40 agonists.

Changyou Zhou; Cheng Tang; Eric Chang; Min Ge; Songnian Lin; Eric Cline; Carina P. Tan; Yue Feng; Yun-Ping Zhou; George J. Eiermann; Aleksandr Petrov; Gino Salituro; Peter T. Meinke; Ralph T. Mosley; Taro E. Akiyama; Monica Einstein; Sanjeev Kumar; Joel P. Berger; Andrew D. Howard; Nancy A. Thornberry; Sander G. Mills; Lihu Yang

Systematic structure-activity relationship (SAR) studies of a screening lead led to the discovery of a series of thiazolidinediones (TZDs) as potent GPR40 agonists. Among them, compound C demonstrated an acute mechanism-based glucose-lowering in an intraperitoneal glucose tolerance test (IPGTT) in lean mice, while no effects were observed in GPR40 knock-out mice.


Journal of Medicinal Chemistry | 2008

Discovery of Potent and Selective Dipeptidyl Peptidase IV Inhibitors Derived from [beta]-Aminoamides Bearing Subsituted Triazolopiperazines

Dooseop Kim; Jennifer E. Kowalchick; Linda Brockunier; Emma R. Parmee; George J. Eiermann; Michael H. Fisher; Huaibing He; Barbara Leiting; Kathryn A. Lyons; Giovanna Scapin; Sangita B. Patel; Aleksandr Petrov; KellyAnn D. Pryor; Ranabir Sinha Roy; Joseph K. Wu; Xiaoping Zhang; Matthew J. Wyvratt; Bei B. Zhang; Lan Zhu; Nancy A. Thornberry; Ann E. Weber

A series of beta-aminoamides bearing triazolopiperazines have been discovered as potent, selective, and orally active dipeptidyl peptidase IV (DPP-4) inhibitors by extensive structure-activity relationship (SAR) studies around the triazolopiperazine moiety. Among these, compound 34b with excellent in vitro potency (IC50 = 4.3 nM) against DPP-4, high selectivity over other enzymes, and good pharmacokinetic profiles exhibited pronounced in vivo efficacy in an oral glucose tolerance test (OGTT) in lean mice. On the basis of these properties, compound 34b has been profiled in detail. Further refinement of the triazolopiperazines resulted in the discovery of a series of extremely potent compounds with subnanomolar activity against DPP-4 (42b- 49b), that is, 4-fluorobenzyl-substituted compound 46b, which is notable for its superior potency (IC50 = 0.18 nM). X-ray crystal structure determination of compounds 34b and 46b in complex with DPP-4 enzyme revealed that (R)-stereochemistry at the 8-position of triazolopiperazines is strongly preferred over (S) with respect to DPP-4 inhibition.


Science | 2017

Systemic pan-AMPK activator MK-8722 improves glucose homeostasis but induces cardiac hypertrophy

Robert W. Myers; Hong-Ping Guan; Juliann Ehrhart; Aleksandr Petrov; Srinivasa Prahalada; Effie Tozzo; Xiaodong Yang; Marc M. Kurtz; Maria E. Trujillo; Dinko Gonzalez Trotter; Danqing Feng; Shiyao Xu; George J. Eiermann; Marie A. Holahan; Daniel Rubins; Stacey Conarello; Xiaoda Niu; Sandra C. Souza; Corin Miller; Jinqi Liu; Ku Lu; Wen Feng; Ying Li; Ronald E. Painter; James A. Milligan; Huaibing He; Franklin Liu; Aimie M. Ogawa; Douglas Wisniewski; Rory J. Rohm

Hitting a dozen enzymes with one drug The adenosine monophosphate-activated protein kinase (AMPK) controls cellular energy status. AMPK is activated when energy levels fall. This stimulates adenosine triphosphate (ATP)-generating pathways that promote glucose uptake and inhibits ATP-consuming pathways associated with glucose synthesis. In principle, these effects would be beneficial in metabolic diseases, including diabetes. Pharmacological activation of AMPK has been challenging, however, because in mammals, the enzyme exists as 12 distinct complexes. Myers et al. describe an orally available compound (MK-8722) that activates all 12 complexes (see the Perspective by Hardie). In animal models, MK-8722 ameliorated diabetes, but it also caused enlargement of the heart. MK-8722 may be a useful tool compound for laboratory research on AMPK function. Science, this issue p. 507; see also p. 455 In animals, a drug activating all 12 isoforms of the energy regulator AMPK benefits metabolism but may pose heart risks. 5′-Adenosine monophosphate–activated protein kinase (AMPK) is a master regulator of energy homeostasis in eukaryotes. Despite three decades of investigation, the biological roles of AMPK and its potential as a drug target remain incompletely understood, largely because of a lack of optimized pharmacological tools. We developed MK-8722, a potent, direct, allosteric activator of all 12 mammalian AMPK complexes. In rodents and rhesus monkeys, MK-8722–mediated AMPK activation in skeletal muscle induced robust, durable, insulin-independent glucose uptake and glycogen synthesis, with resultant improvements in glycemia and no evidence of hypoglycemia. These effects translated across species, including diabetic rhesus monkeys, but manifested with concomitant cardiac hypertrophy and increased cardiac glycogen without apparent functional sequelae.


Journal of Pharmacology and Experimental Therapeutics | 2013

The role of voltage-gated potassium channels Kv2.1 and Kv2.2 in the regulation of insulin and somatostatin release from pancreatic islets.

Xiaoyan Li; James Herrington; Aleksandr Petrov; Lan Ge; George J. Eiermann; Yusheng Xiong; Mette V. Jensen; Hans E. Hohmeier; Christopher B. Newgard; Maria L. Garcia; Michael Wagner; Bei B. Zhang; Nancy A. Thornberry; Andrew D. Howard; Gregory J. Kaczorowski; Yun-Ping Zhou

The voltage-gated potassium channels Kv2.1 and Kv2.2 are highly expressed in pancreatic islets, yet their contribution to islet hormone secretion is not fully understood. Here we investigate the role of Kv2 channels in pancreatic islets using a combination of genetic and pharmacologic approaches. Pancreatic β-cells from Kv2.1−/− mice possess reduced Kv current and display greater glucose-stimulated insulin secretion (GSIS) relative to WT β-cells. Inhibition of Kv2.x channels with selective peptidyl [guangxitoxin-1E (GxTX-1E)] or small molecule (RY796) inhibitors enhances GSIS in isolated wild-type (WT) mouse and human islets, but not in islets from Kv2.1−/− mice. However, in WT mice neither inhibitor improved glucose tolerance in vivo. GxTX-1E and RY796 enhanced somatostatin release in isolated human and mouse islets and in situ perfused pancreata from WT and Kv2.1−/− mice. Kv2.2 silencing in mouse islets by adenovirus-small hairpin RNA (shRNA) specifically enhanced islet somatostatin, but not insulin, secretion. In mice lacking somatostatin receptor 5, GxTX-1E stimulated insulin secretion and improved glucose tolerance. Collectively, these data show that Kv2.1 regulates insulin secretion in β-cells and Kv2.2 modulates somatostatin release in δ-cells. Development of selective Kv2.1 inhibitors without cross inhibition of Kv2.2 may provide new avenues to promote GSIS for the treatment of type 2 diabetes.


American Journal of Physiology-endocrinology and Metabolism | 2012

Differential effects of oxyntomodulin and GLP-1 on glucose metabolism

Xiaobing Du; Jennifer R. Kosinski; Julie Lao; Xiaolan Shen; Aleksandr Petrov; Gary G. Chicchi; George J. Eiermann; Alessandro Pocai

Glucagon-like peptide-1 (GLP-1) and oxyntomodulin (OXM) are peptide hormones secreted postprandially from the gut that stimulate insulin secretion in a glucose-dependent manner. OXM activates both the GLP-1 receptor (GLP1R) and the glucagon receptor (GCGR). It has been suggested that OXM acutely modulates glucose metabolism solely through GLP1R agonism. Because OXM activates the GLP1R with lower affinity than GLP-1, we generated a peptide analog (Q→E, OXMQ3E) that does not exhibit glucagon receptor agonist activity but retains the same affinity as OXM for GLP1R. We compared the effects of OXM and OXMQ3E in a glucose tolerance test and, to better characterize the effect on glucose metabolism, we performed controlled infusions of OXM or OXMQ3E during a hyperglycemic clamp performed in wild-type, Glp1r(-/-), and Gcgr(-/-) mice. Our findings show that OXM, but not OXMQ3E, activates the GCGR in vivo. Second, OXM and OXMQ3E improve glucose tolerance following an acute glucose challenge and during a hyperglycemic clamp in mice. Finally, OXM infusion during a glucose clamp reduces the glucose infusion rate (GIR) despite a simultaneous increase in insulin levels in Glp1r(-/-) mice, whereas OXM and OXMQ3E increase GIR to a similar extent in Gcgr(-/-) mice. In conclusion, activation of the GCGR seems to partially attenuate the acute beneficial effects on glucose and contributes to the insulinotropic action of oxyntomodulin.

Collaboration


Dive into the Aleksandr Petrov's collaboration.

Researchain Logo
Decentralizing Knowledge