Network


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

Hotspot


Dive into the research topics where Julie S. Moyers is active.

Publication


Featured researches published by Julie S. Moyers.


Journal of Clinical Investigation | 2005

FGF-21 as a novel metabolic regulator

Alexei Kharitonenkov; Tatiyana L. Shiyanova; Anja Koester; Amy M. Ford; Radmila Micanovic; Elizabeth Galbreath; George E. Sandusky; Lisa Janine Hammond; Julie S. Moyers; Rebecca A. Owens; Jesper Gromada; Joseph T. Brozinick; Eric Hawkins; Victor J. Wroblewski; De Shan Li; Farrokh Mehrbod; S. Richard Jaskunas; Armen B. Shanafelt

Diabetes mellitus is a major health concern, affecting more than 5% of the population. Here we describe a potential novel therapeutic agent for this disease, FGF-21, which was discovered to be a potent regulator of glucose uptake in mouse 3T3-L1 and primary human adipocytes. FGF-21-transgenic mice were viable and resistant to diet-induced obesity. Therapeutic administration of FGF-21 reduced plasma glucose and triglycerides to near normal levels in both ob/ob and db/db mice. These effects persisted for at least 24 hours following the cessation of FGF-21 administration. Importantly, FGF-21 did not induce mitogenicity, hypoglycemia, or weight gain at any dose tested in diabetic or healthy animals or when overexpressed in transgenic mice. Thus, we conclude that FGF-21, which we have identified as a novel metabolic factor, exhibits the therapeutic characteristics necessary for an effective treatment of diabetes.


Journal of Cellular Physiology | 2008

FGF-21/FGF-21 receptor interaction and activation is determined by βKlotho

Alexei Kharitonenkov; James D. Dunbar; Holly A. Bina; Stuart Willis Bright; Julie S. Moyers; Chen Zhang; Liyun Ding; Radmila Micanovic; Sean F. Mehrbod; Michael D. Knierman; John E. Hale; Tamer Coskun; Armen B. Shanafelt

Fibroblast growth factor‐21 (FGF‐21) is a metabolic regulator that can influence glucose and lipid control in diabetic rodents and primates. We demonstrate that βKlotho is an integral part of an activated FGF‐21‐βKlotho‐FGF receptor (FGFR) complex thus a critical subunit of the FGF‐21 receptor. Cells lacking βKlotho did not respond to FGF‐21; the introduction of βKlotho to these cells conferred FGF‐21‐responsiveness and recapitulated the entire scope of FGF‐21 signaling observed in naturally responsive cells. Interestingly, FGF‐21‐mediated effects are heparin independent suggesting that βKlotho plays a role in FGF‐21 activity similar to the one played by heparin in the signaling of conventional FGFs. Moreover, in addition to conferring specificity for FGF‐21, βKlotho appears to support FGF‐19 activity and mediates the receptor selectivity profile of FGF‐19. All together, these results indicate that βKlotho and FGFRs form the cognate FGF‐21 receptor complex, mediating FGF‐21 cellular specificity and physiological effects. J. Cell. Physiol. 215: 1–7, 2008.


Journal of Cellular Physiology | 2007

Molecular determinants of FGF-21 activity : Synergy and cross-talk with PPARγ signaling

Julie S. Moyers; Tatiyana L. Shiyanova; Farrokh Mehrbod; James D. Dunbar; Timothy Wayne Noblitt; Keith A. Otto; Anne Reifel-Miller; Alexei Kharitonenkov

Fibroblast growth factor (FGF)‐21 is a novel regulator of insulin‐independent glucose transport in 3T3‐L1 adipocytes and has glucose and triglyceride lowering effects in rodent models of diabetes. The precise mechanisms whereby FGF‐21 regulates metabolism remain to be determined. Here we describe the early signaling events triggered by FGF‐21 treatment of 3T3‐L1 adipocytes and reveal a functional interplay between FGF‐21 and peroxisome proliferator‐activated receptor gamma (PPARγ) pathways that leads to a marked stimulation of glucose transport. While the early actions of FGF‐21 on 3T3‐L1 adipocytes involve rapid accumulation of intracellular calcium and phosphorylation of Akt, GSK‐3, p70S6K, SHP‐2, MEK1/2, and Stat3, continuous treatment for 72 h induces an increase in PPARγ protein expression. Moreover, chronic activation of the PPARγ pathway in 3T3‐L1 adipocytes with the PPARγ agonist and anti‐diabetic agent, rosiglitazone (BRL 49653), enhances FGF‐21 action to induce tyrosine phosphorylation of FGF receptor‐2. Strikingly, treatment of cells with FGF‐21 and rosiglitazone in combination leads to a pronounced increase in expression of the GLUT1 glucose transporter and a marked synergy in stimulation of glucose transport. Together these results reveal a novel synergy between two regulators of glucose homeostasis, FGF‐21 and PPARγ, and further define FGF‐21 mechanism of action. J. Cell. Physiol. 210: 1–6, 2007.


Expert Opinion on Therapeutic Targets | 2005

Glucagon as a target for the treatment of Type 2 diabetes.

Kyle W. Sloop; M. Dodson Michael; Julie S. Moyers

Glucagon is the key counter-regulatory hormone that opposes the action of insulin. In states of relative hypoglycaemia, glucagon acts to increase blood glucose by stimulating hepatic glycogen breakdown and gluconeogenesis to achieve euglycaemia. Type 2 diabetes is characterised by inappropriate regulation of hepatic glucose production, which is due, at least in part, to an imbalance in the bihormonal relationship between plasma levels of glucagon and insulin. The glucose-lowering effects of glucagon peptide antagonists and antiglucagon neutralising antibodies first demonstrated the potential of glucagon receptor (GCGR) antagonism as a treatment for hyperglycaemia. In recent years, the development of GCGR antisense oligonucleotides and small molecular weight GCGR antagonists have been pursued as possible therapeu-tic agents to target glucagon action as a treatment for Type 2 diabe-tes.


eLife | 2016

Blockade of glucagon signaling prevents or reverses diabetes onset only if residual β-cells persist

Nicolas Damond; Fabrizio Thorel; Julie S. Moyers; Maureen J. Charron; Patricia M Vuguin; Alvin C. Powers; Pedro Luis Herrera

Glucagon secretion dysregulation in diabetes fosters hyperglycemia. Recent studies report that mice lacking glucagon receptor (Gcgr-/-) do not develop diabetes following streptozotocin (STZ)-mediated ablation of insulin-producing β-cells. Here, we show that diabetes prevention in STZ-treated Gcgr-/- animals requires remnant insulin action originating from spared residual β-cells: these mice indeed became hyperglycemic after insulin receptor blockade. Accordingly, Gcgr-/- mice developed hyperglycemia after induction of a more complete, diphtheria toxin (DT)-induced β-cell loss, a situation of near-absolute insulin deficiency similar to type 1 diabetes. In addition, glucagon deficiency did not impair the natural capacity of α-cells to reprogram into insulin production after extreme β-cell loss. α-to-β-cell conversion was improved in Gcgr-/- mice as a consequence of α-cell hyperplasia. Collectively, these results indicate that glucagon antagonism could i) be a useful adjuvant therapy in diabetes only when residual insulin action persists, and ii) help devising future β-cell regeneration therapies relying upon α-cell reprogramming. DOI: http://dx.doi.org/10.7554/eLife.13828.001


Journal of Pharmacology and Experimental Therapeutics | 2016

In Vivo and In Vitro Characterization of Basal Insulin Peglispro: A Novel Insulin Analog

Rebecca A. Owens; Ryan John Hansen; Steven D. Kahl; Chen Zhang; Xiaoping Ruan; Anja Koester; Shun Li; Hui-Rong Qian; Mark W Farman; Dodson Michael; Julie S. Moyers; Gordon B. Cutler; Andrew Mark Vick; John Michael Beals

The aim of this research was to characterize the in vivo and in vitro properties of basal insulin peglispro (BIL), a new basal insulin, wherein insulin lispro was derivatized through the covalent and site-specific attachment of a 20-kDa polyethylene-glycol (PEG; specifically, methoxy-terminated) moiety to lysine B28. Addition of the PEG moiety increased the hydrodynamic size of the insulin lispro molecule. Studies show there is a prolonged duration of action and a reduction in clearance. Given the different physical properties of BIL, it was also important to assess the metabolic and mitogenic activity of the molecule. Streptozotocin (STZ)-treated diabetic rats were used to study the pharmacokinetic and pharmacodynamic characteristics of BIL. Binding affinity and functional characterization of BIL were compared with those of several therapeutic insulins, insulin AspB10, and insulin-like growth factor 1 (IGF-1). BIL exhibited a markedly longer time to maximum concentration after subcutaneous injection, a greater area under the concentration-time curve, and a longer duration of action in the STZ-treated diabetic rat than insulin lispro. BIL exhibited reduced binding affinity and functional potency as compared with insulin lispro and demonstrated greater selectivity for the human insulin receptor (hIR) as compared with the human insulin-like growth factor 1 receptor. Furthermore, BIL showed a more rapid rate of dephosphorylation following maximal hIR stimulation, and reduced mitogenic potential in an IGF-1 receptor–dominant cellular model. PEGylation of insulin lispro with a 20-kDa PEG moiety at lysine B28 alters the absorption, clearance, distribution, and activity profile receptor, but does not alter its selectivity and full agonist receptor properties.


Regulatory Toxicology and Pharmacology | 2017

Nonclinical pharmacology and toxicology of the first biosimilar insulin glargine drug product (BASAGLAR®/ABASAGLAR®) approved in the European Union

Richard A. Byrd; Rebecca A. Owens; Jamie L. Blackbourne; David E. Coutant; Mark W. Farmen; M. Dodson Michael; Julie S. Moyers; A. Eric Schultze; Michael K. Sievert; Niraj K. Tripathi; John L. Vahle

ABSTRACT Basaglar®/Abasaglar® (Lilly insulin glargine [LY IGlar]) is a long‐acting human insulin analogue drug product granted marketing authorisation as a biosimilar to Lantus® (Sanofi insulin glargine [SA IGlar]) by the European Medicines Agency. We assessed the similarity of LY IGlar to the reference drug product, European Union–sourced SA IGlar (EU‐SA IGlar), using nonclinical in vitro and in vivo studies. No biologically relevant differences were observed for receptor binding affinity at either the insulin or insulin‐like growth factor‐1 (IGF‐1) receptors, or in assays of functional or de novo lipogenic activity. The mitogenic potential of LY IGlar and EU‐SA IGlar was similar when tested in both insulin‐ and IGF‐1 receptor dominant cell systems. Repeated subcutaneous daily dosing of rats for 4 weeks with 0, 0.3, 1.0, or 2.0 mg/kg LY IGlar and EU‐SA IGlar produced mortalities and clinical signs consistent with severe hypoglycaemia. Glucodynamic profiles of LY IGlar and EU‐SA IGlar in satellite animals showed comparable dose‐related hypoglycaemia. Severe hypoglycaemia was associated with axonal degeneration of the sciatic nerve; the incidence and severity were low and did not differ between LY IGlar and EU‐SA IGlar. These results demonstrated no biologically relevant differences in toxicity between LY IGlar and EU‐SA IGlar. HIGHLIGHTSBasaglar®/Abasaglar® (LY IGlar) is the first biosimilar insulin glargine drug product approved in the European Union.We compared nonclinical profiles of LY IGlar and European Union–sourced Lantus (EU‐SA IGlar).We found no biologically relevant differences between LY IGlar and EU‐SA IGlar.


Molecular metabolism | 2018

LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: From discovery to clinical proof of concept

Tamer Coskun; Kyle W. Sloop; Corina Loghin; Jorge Alsina-Fernandez; Shweta Urva; Krister Bokvist; Xuewei Cui; Daniel A. Briere; Over Cabrera; William C. Roell; Uma Kuchibhotla; Julie S. Moyers; Charles Benson; Ruth E. Gimeno; David A. D'Alessio; Axel Haupt

Objective A novel dual GIP and GLP-1 receptor agonist, LY3298176, was developed to determine whether the metabolic action of GIP adds to the established clinical benefits of selective GLP-1 receptor agonists in type 2 diabetes mellitus (T2DM). Methods LY3298176 is a fatty acid modified peptide with dual GIP and GLP-1 receptor agonist activity designed for once-weekly subcutaneous administration. LY3298176 was characterised in vitro, using signaling and functional assays in cell lines expressing recombinant or endogenous incretin receptors, and in vivo using body weight, food intake, insulin secretion and glycemic profiles in mice. A Phase 1, randomised, placebo-controlled, double-blind study was comprised of three parts: a single-ascending dose (SAD; doses 0.25–8 mg) and 4-week multiple-ascending dose (MAD; doses 0.5–10 mg) studies in healthy subjects (HS), followed by a 4-week multiple-dose Phase 1 b proof-of-concept (POC; doses 0.5–15 mg) in patients with T2DM (ClinicalTrials.gov no. NCT02759107). Doses higher than 5 mg were attained by titration, dulaglutide (DU) was used as a positive control. The primary objective was to investigate safety and tolerability of LY3298176. Results LY3298176 activated both GIP and GLP-1 receptor signaling in vitro and showed glucose-dependent insulin secretion and improved glucose tolerance by acting on both GIP and GLP-1 receptors in mice. With chronic administration to mice, LY3298176 potently decreased body weight and food intake; these effects were significantly greater than the effects of a GLP-1 receptor agonist. A total of 142 human subjects received at least 1 dose of LY3298176, dulaglutide, or placebo. The PK profile of LY3298176 was investigated over a wide dose range (0.25–15 mg) and supports once-weekly administration. In the Phase 1 b trial of diabetic subjects, LY3298176 doses of 10 mg and 15 mg significantly reduced fasting serum glucose compared to placebo (least square mean [LSM] difference [95% CI]: −49.12 mg/dL [−78.14, −20.12] and −43.15 mg/dL [−73.06, −13.21], respectively). Reductions in body weight were significantly greater with the LY3298176 1.5 mg, 4.5 mg and 10 mg doses versus placebo in MAD HS (LSM difference [95% CI]: −1.75 kg [−3.38, −0.12], −5.09 kg [−6.72, −3.46] and −4.61 kg [−6.21, −3.01], respectively) and doses of 10 mg and 15 mg had a relevant effect in T2DM patients (LSM difference [95% CI]: −2.62 kg [−3.79, −1.45] and −2.07 kg [−3.25, −0.88], respectively. The most frequent side effects reported with LY3298176 were gastrointestinal (vomiting, nausea, decreased appetite, diarrhoea, and abdominal distension) in both HS and patients with T2DM; all were dose-dependent and considered mild to moderate in severity. Conclusions Based on these results, the pharmacology of LY3298176 translates from preclinical to clinical studies. LY3298176 has the potential to deliver clinically meaningful improvement in glycaemic control and body weight. The data warrant further clinical evaluation of LY3298176 for the treatment of T2DM and potentially obesity.


Diabetes | 2018

Insulin-XTEN® Exhibits a Size-Dependent Alteration in Tissue Action in Rats

Michael E. Christe; Debra L. Konkol; Jessica Friedrich; J. R. Jacobs; Eric Hawkins; Julie S. Moyers; Chen Zhang; Steven D. Kahl; Hana E. Baker; Amy L. Cox; Ryan John Hansen; Andrea Sperry; M. Dodson Michael; Volker Schellenberger; D. Bruce Baldwin; John Michael Beals; Andrew Ihor Korytko

To optimize the action of exogenously administered insulin, we employed XTEN® technology to create insulins with variably sized XTEN amino acid polymers. Recombinant fusions of XTEN polymers linked to insulin lispro with an A21G mutation were prepared in various amino acid lengths. Insulin-XTEN molecules demonstrated 15-fold lower potency in binding and receptor phosphorylation than insulin lispro but did not differ from each other. These insulin-XTEN molecules were equally effective in lowering blood glucose at a 100nmol/kg dose in diabetic Sprague-Dawley rats. Furthermore, the larger insulin-XTEN molecules had a longer duration of glucose lowering. Insulin-XTENs were compared to insulin lispro in rat euglycemic clamp studies, using insulin doses that would elicit steady plasma insulin concentrations and equivalent increases in glucose infusion rate. Insulin-mediated suppression of endogenous glucose production was not significantly different among any of the administered insulins. However, plasma free fatty acids and soleus muscle glucose uptake were significantly decreased in an XTEN size-dependent manner when compared to insulin lispro. Additional studies demonstrated equal hepatic pAkt accumulation in rats treated with insulin lispro or any of the insulin-XTENs, but revealed a significant XTEN size-dependent reduction in skeletal muscle pAkt in rats administered insulin-XTENs compared to insulin lispro. These data suggest a possible XTEN size-dependent regulation of insulin action and that the differing sizes of the XTEN polymer may convey preferential tissue action. In conclusion, XTEN technology may permit “tuning” of the glucodynamic effects of the insulin, leading to an enhanced time extension and improved hepatic and peripheral pharmacodynamic action that could more closely mimic the action of endogenously secreted insulin into the portal circulation. Disclosure M.E. Christe: Employee; Self; Eli Lilly and Company. D. Konkol: None. J. Friedrich: None. J. Jacobs: None. E. Hawkins: Employee; Self; Eli Lilly and Company. J. Moyers: Employee; Self; Eli Lilly and Company. Stock/Shareholder; Self; Eli Lilly and Company. C. Zhang: Employee; Self; Eli Lilly and Company. S.D. Kahl: Employee; Self; Eli Lilly and Company. H.E. Baker: None. A.L. Cox: None. R.J. Hansen: Employee; Self; Eli Lilly and Company. Stock/Shareholder; Self; Eli Lilly and Company. A. Sperry: Employee; Self; Eli Lilly and Company. Stock/Shareholder; Self; Eli Lilly and Company. M. Michael: Employee; Self; Eli Lilly and Company. Stock/Shareholder; Self; Eli Lilly and Company. Employee; Spouse/Partner; Eli Lilly and Company. Stock/Shareholder; Spouse/Partner; Eli Lilly and Company. V. Schellenberger: None. D. Baldwin: None. J.M. Beals: Employee; Self; Eli Lilly and Company. A. Korytko: None.


Journal of Clinical Investigation | 2004

Hepatic and glucagon-like peptide-1–mediated reversal of diabetes by glucagon receptor antisense oligonucleotide inhibitors

Kyle W. Sloop; Julia Xiao-Chun Cao; Angela M. Siesky; Hong Yan Zhang; Diane M. Bodenmiller; Amy L. Cox; Steven J. Jacobs; Julie S. Moyers; Rebecca A. Owens; Aaron D. Showalter; Martin B. Brenner; Achim Raap; Jesper Gromada; Brian R. Berridge; David Monteith; Niels Porksen; Robert Mckay; Brett P. Monia; Sanjay Bhanot; Lynnetta Watts; M. Dodson Michael

Collaboration


Dive into the Julie S. Moyers's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge