Network


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

Hotspot


Dive into the research topics where Nickki Ottaway is active.

Publication


Featured researches published by Nickki Ottaway.


Nature Chemical Biology | 2009

A new glucagon and GLP-1 co-agonist eliminates obesity in rodents

Jonathan Day; Nickki Ottaway; James T. Patterson; Vasily Gelfanov; David L. Smiley; Jas Gidda; Hannes M. Findeisen; Dennis Bruemmer; Daniel J. Drucker; Nilika Chaudhary; Jenna Holland; Jazzminn Hembree; William Abplanalp; Erin Grant; Jennifer Ruehl; Hilary Wilson; Henriette Kirchner; Sarah Kathleen Haas Lockie; Susanna M. Hofmann; Stephen C. Woods; Ruben Nogueiras; Paul T. Pfluger; Diego Perez-Tilve; Richard D. DiMarchi; Matthias H. Tschöp

We report the efficacy of a new peptide with agonism at the glucagon and GLP-1 receptors that has potent, sustained satiation-inducing and lipolytic effects. Selective chemical modification to glucagon resulted in a loss of specificity, with minimal change to inherent activity. The structural basis for the co-agonism appears to be a combination of local positional interactions and a change in secondary structure. Two co-agonist peptides differing from each other only in their level of glucagon receptor agonism were studied in rodent obesity models. Administration of PEGylated peptides once per week normalized adiposity and glucose tolerance in diet-induced obese mice. Reduction of body weight was achieved by a loss of body fat resulting from decreased food intake and increased energy expenditure. These preclinical studies indicate that when full GLP-1 agonism is augmented with an appropriate degree of glucagon receptor activation, body fat reduction can be substantially enhanced without any overt adverse effects.


Science Translational Medicine | 2013

Unimolecular Dual Incretins Maximize Metabolic Benefits in Rodents, Monkeys, and Humans

Brian Finan; Tao Ma; Nickki Ottaway; Timo D. Müller; Kirk M. Habegger; Kristy M. Heppner; Henriette Kirchner; Jenna Holland; Jazzminn Hembree; Christine Raver; Sarah Kathleen Haas Lockie; David L. Smiley; Vasily Gelfanov; Bin Yang; Susanna M. Hofmann; Dennis Bruemmer; Daniel J. Drucker; Paul T. Pfluger; Diego Perez-Tilve; Jaswant Gidda; Louis Vignati; Lianshan Zhang; Jonathan Hauptman; Michele Lau; Mathieu Brecheisen; Sabine Uhles; William Riboulet; Emmanuelle Hainaut; Elena Sebokova; Karin Conde-Knape

Compared to best-in-class GLP-1 mono-agonists, unimolecular co-agonists of GLP-1 and GIP with optimized pharmacokinetics enhance glycemic and metabolic benefits in mammals. “Twincretins”: Two Is Better than One Despite obesity-linked diabetes approaching worldwide epidemic proportions and the growing recognition of it as a global health challenge, safe and effective medicines have remained largely elusive. Pharmacological options targeting multiple obesity and diabetes signaling pathways offer greater therapeutic potential compared to molecules targeting a single pathway. Finan et al. now report the discovery, characterization, and translational efficacy of a single molecule that acts equally on the receptors for the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). In rodent models of obesity and diabetes, this dual incretin co-agonist more effectively lowered body fat and corrected hyperglycemia than selective mono-agonists for the GLP-1 and GIP receptors. An enhanced insulinotropic effect translated from rodents to monkeys and humans, with substantially improved levels of glycosylated hemoglobin A1c (HbA1c) in humans with type 2 diabetes. The dual incretin was engineered with selective chemical modifications to enhance pharmacokinetics. This, in combination with its inherent mixed agonism, lowered the drug dose and ameliorated the dose-limiting nausea that has plagued selective GLP-1 therapies. These dual incretin co-agonists signify a new direction for unimolecular combination therapy and represent a new class of drug candidates for the treatment of metabolic diseases. We report the discovery and translational therapeutic efficacy of a peptide with potent, balanced co-agonism at both of the receptors for the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). This unimolecular dual incretin is derived from an intermixed sequence of GLP-1 and GIP, and demonstrated enhanced antihyperglycemic and insulinotropic efficacy relative to selective GLP-1 agonists. Notably, this superior efficacy translated across rodent models of obesity and diabetes, including db/db mice and ZDF rats, to primates (cynomolgus monkeys and humans). Furthermore, this co-agonist exhibited synergism in reducing fat mass in obese rodents, whereas a selective GIP agonist demonstrated negligible weight-lowering efficacy. The unimolecular dual incretins corrected two causal mechanisms of diabesity, adiposity-induced insulin resistance and pancreatic insulin deficiency, more effectively than did selective mono-agonists. The duration of action of the unimolecular dual incretins was refined through site-specific lipidation or PEGylation to support less frequent administration. These peptides provide comparable pharmacology to the native peptides and enhanced efficacy relative to similarly modified selective GLP-1 agonists. The pharmacokinetic enhancement lessened peak drug exposure and, in combination with less dependence on GLP-1–mediated pharmacology, avoided the adverse gastrointestinal effects that typify selective GLP-1–based agonists. This discovery and validation of a balanced and high-potency dual incretin agonist enables a more physiological approach to management of diseases associated with impaired glucose tolerance.


Nature Medicine | 2015

A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents

Brian Finan; Bin Yang; Nickki Ottaway; David L. Smiley; Tao Ma; Christoffer Clemmensen; Joe Chabenne; Lianshan Zhang; Kirk M. Habegger; Katrin Fischer; Jonathan E. Campbell; Darleen A. Sandoval; Randy J. Seeley; Konrad Bleicher; Sabine Uhles; William Riboulet; Jürgen Funk; Cornelia Hertel; Sara Belli; Elena Sebokova; Karin Conde-Knape; Anish Konkar; Daniel J. Drucker; Vasily Gelfanov; Paul T. Pfluger; Timo D. Müller; Diego Perez-Tilve; Richard D. DiMarchi; Matthias H. Tschöp

We report the discovery of a new monomeric peptide that reduces body weight and diabetic complications in rodent models of obesity by acting as an agonist at three key metabolically-related peptide hormone receptors: glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP) and glucagon receptors. This triple agonist demonstrates supraphysiological potency and equally aligned constituent activities at each receptor, all without cross-reactivity at other related receptors. Such balanced unimolecular triple agonism proved superior to any existing dual coagonists and best-in-class monoagonists to reduce body weight, enhance glycemic control and reverse hepatic steatosis in relevant rodent models. Various loss-of-function models, including genetic knockout, pharmacological blockade and selective chemical knockout, confirmed contributions of each constituent activity in vivo. We demonstrate that these individual constituent activities harmonize to govern the overall metabolic efficacy, which predominantly results from synergistic glucagon action to increase energy expenditure, GLP-1 action to reduce caloric intake and improve glucose control, and GIP action to potentiate the incretin effect and buffer against the diabetogenic effect of inherent glucagon activity. These preclinical studies suggest that, so far, this unimolecular, polypharmaceutical strategy has potential to be the most effective pharmacological approach to reversing obesity and related metabolic disorders.


Nature Medicine | 2012

Targeted estrogen delivery reverses the metabolic syndrome

Brian Finan; Bin Yang; Nickki Ottaway; Kerstin Stemmer; Timo D. Müller; Chun Xia Yi; Kirk M. Habegger; Sonja C. Schriever; Cristina García-Cáceres; Dhiraj G. Kabra; Jazzminn Hembree; Jenna Holland; Christine Raver; Randy J. Seeley; Wolfgang Hans; Martin Irmler; Johannes Beckers; Martin Hrabě de Angelis; Joseph P. Tiano; Franck Mauvais-Jarvis; Diego Perez-Tilve; Paul T. Pfluger; Lianshan Zhang; Vasily Gelfanov; Richard D. DiMarchi; Matthias H. Tschöp

We report the development of a new combinatorial approach that allows for peptide-mediated selective tissue targeting of nuclear hormone pharmacology while eliminating adverse effects in other tissues. Specifically, we report the development of a glucagon-like peptide-1 (GLP-1)-estrogen conjugate that has superior sex-independent efficacy over either of the individual hormones alone to correct obesity, hyperglycemia and dyslipidemia in mice. The therapeutic benefits are driven by pleiotropic dual hormone action to improve energy, glucose and lipid metabolism, as shown by loss-of-function models and genetic action profiling. Notably, the peptide-based targeting strategy also prevents hallmark side effects of estrogen in male and female mice, such as reproductive endocrine toxicity and oncogenicity. Collectively, selective activation of estrogen receptors in GLP-1–targeted tissues produces unprecedented efficacy to enhance the metabolic benefits of GLP-1 agonism. This example of targeting the metabolic syndrome represents the discovery of a new class of therapeutics that enables synergistic co-agonism through peptide-based selective delivery of small molecules. Although our observations with the GLP-1–estrogen conjugate justify translational studies for diabetes and obesity, the multitude of other possible combinations of peptides and small molecules may offer equal promise for other diseases.


Diabetes | 2013

Fibroblast Growth Factor 21 Mediates Specific Glucagon Actions

Kirk M. Habegger; Kerstin Stemmer; Christine C. Cheng; Timo D. Müller; Kristy M. Heppner; Nickki Ottaway; Jenna Holland; Jazzminn Hembree; David L. Smiley; Vasily Gelfanov; Radha Krishna; Ayman M. Arafat; Anish Konkar; Sara Belli; Martin Kapps; Stephen C. Woods; Susanna M. Hofmann; David A. D’Alessio; Paul T. Pfluger; Diego Perez-Tilve; Randy J. Seeley; Morichika Konishi; Nobuyujki Itoh; Alexei Kharitonenkov; Joachim Spranger; Richard D. DiMarchi; Matthias H. Tschöp

Glucagon, an essential regulator of glucose homeostasis, also modulates lipid metabolism and promotes weight loss, as reflected by the wasting observed in glucagonoma patients. Recently, coagonist peptides that include glucagon agonism have emerged as promising therapeutic candidates for the treatment of obesity and diabetes. We developed a novel stable and soluble glucagon receptor (GcgR) agonist, which allowed for in vivo dissection of glucagon action. As expected, chronic GcgR agonism in mice resulted in hyperglycemia and lower body fat and plasma cholesterol. Notably, GcgR activation also raised hepatic expression and circulating levels of fibroblast growth factor 21 (FGF21). This effect was retained in isolated primary hepatocytes from wild-type (WT) mice, but not GcgR knockout mice. We confirmed this link in healthy human volunteers, where injection of natural glucagon increased plasma FGF21 within hours. Functional relevance was evidenced in mice with genetic deletion of FGF21, where GcgR activation failed to induce the body weight loss and lipid metabolism changes observed in WT mice. Taken together, these data reveal for the first time that glucagon controls glucose, energy, and lipid metabolism at least in part via FGF21-dependent pathways.


Journal of Peptide Science | 2012

Restoration of leptin responsiveness in diet-induced obese mice using an optimized leptin analog in combination with exendin-4 or FGF21.

Timo D. Müller; Lorraine Sullivan; Kirk M. Habegger; Chun-Xia Yi; Dhiraj G. Kabra; Erin Grant; Nickki Ottaway; Radha Krishna; Jenna Holland; Jazzminn Hembree; Diego Perez-Tilve; Paul T. Pfluger; Michael J. DeGuzman; Marc E. Siladi; Douglas W. Axelrod; Richard D. DiMarchi; Jason Pinkstaff; Matthias H. Tschöp

The identification of leptin as a mediator of body weight regulation provided much initial excitement for the treatment of obesity. Unfortunately, leptin monotherapy is insufficient in reversing obesity in rodents or humans. Recent findings suggest that amylin is able to restore leptin sensitivity and when used in combination with leptin enhances body weight loss in obese rodents and humans. However, as the uniqueness of this combination therapy remains unclear, we assessed whether co‐administration of leptin with other weight loss‐inducing hormones equally restores leptin responsiveness in diet‐induced obese (DIO) mice. Accordingly, we report here the design and characterization of a series of site‐specifically enhanced leptin analogs of high potency and sustained action that, when administered in combination with exendin‐4 or fibroblast growth factor 21 (FGF21), restores leptin responsiveness in DIO mice after an initial body weight loss of 30%. Using either combination, body weight loss was enhanced compared with either exendin‐4 or FGF21 monotherapy, and leptin alone was sufficient to maintain the reduced body weight. In contrast, leptin monotherapy proved ineffective when identical weight loss was induced by caloric restriction alone over a comparable time. Accordingly, we find that a hypothalamic counter‐regulatory response to weight loss, assessed using changes in hypothalamic agouti related peptide (AgRP) levels, is triggered by caloric restriction, but blunted by treatment with exendin‐4. We conclude that leptin re‐sensitization requires pharmacotherapy but does not appear to be restricted to a unique signaling pathway. Our findings provide preclinical evidence that high activity, long‐acting leptin analogs are additively efficacious when used in combination with other weight‐lowering agents. Copyright


Glia | 2014

Hormones and diet, but not body weight, control hypothalamic microglial activity.

Yuanqing Gao; Nickki Ottaway; Sonja C. Schriever; Beata Legutko; Cristina García-Cáceres; Esther de la Fuente; Clarita Mergen; Susanne Bour; Joshua P. Thaler; Randy J. Seeley; Jessica A. Filosa; Javier E. Stern; Diego Perez-Tilve; Michael W. Schwartz; Matthias H. Tschöp; Chun Xia Yi

The arcuate nucleus (ARC) of the hypothalamus plays a key role in sensing metabolic feedback and regulating energy homeostasis. Recent studies revealed activation of microglia in mice with high‐fat diet (HFD)‐induced obesity (DIO), suggesting a potential pathophysiological role for inflammatory processes within the hypothalamus. To further investigate the metabolic causes and molecular underpinnings of such glial activation, we analyzed the microglial activity in wild‐type (WT), monogenic obese ob/ob (leptin deficient), db/db (leptin‐receptor mutation), and Type‐4 melanocortin receptor knockout (MC4R KO) mice on either a HFD or on standardized chow (SC) diet. Following HFD exposure, we observed a significant increase in the total number of ARC microglia, immunoreactivity of ionized calcium binding adaptor molecule 1 (iba1‐ir), cluster of differentiation 68 (CD68‐ir), and ramification of microglial processes. The ob/ob mice had significantly less iba1‐ir and ramifications. Leptin replacement rescued these phenomena. The db/db mice had similar iba1‐ir comparable with WT mice but had significantly lower CD68‐ir and more ramifications than WT mice. After 2 weeks of HFD, ob/ob mice showed an increase of iba1‐ir, and db/db mice showed increase of CD68‐ir. Obese MC4R KO mice fed a SC diet had comparable iba1‐ir and CD68‐ir with WT mice but had significantly more ramifications than WT mice. Intriguingly, treatment of DIO mice with glucagon‐like peptide‐1 receptor agonists reduced microglial activation independent of body weight. Our results show that diet type, adipokines, and gut signals, but not body weight, affect the presence and activity levels of hypothalamic microglia in obesity. GLIA 2013;62:17–25


The Journal of Neuroscience | 2010

CNS Leptin Action Modulates Immune Response and Survival in Sepsis

Johannes Tschöp; Ruben Nogueiras; Sarah Haas-Lockie; Kevin R. Kasten; Tamara R. Castañeda; Nadine Huber; Kelsey Guanciale; Diego Perez-Tilve; Kirk M. Habegger; Nickki Ottaway; Stephen C. Woods; Brian J. Oldfield; Iain J. Clarke; Streamson C. Chua; Farooqi Is; Stephen O'Rahilly; Charles C. Caldwell; M. Tschöp

Sepsis describes a complex clinical syndrome that results from an infection, setting off a cascade of systemic inflammatory responses that can lead to multiple organ failure and death. Leptin is a 16 kDa adipokine that, among its multiple known effects, is involved in regulating immune function. Here we demonstrate that leptin deficiency in ob/ob mice leads to higher mortality and more severe organ damage in a standard model of sepsis in mice [cecal ligation and puncture (CLP)]. Moreover, systemic leptin replacement improved the immune response to CLP. Based on the molecular mechanisms of leptin regulation of energy metabolism and reproductive function, we hypothesized that leptin acts in the CNS to efficiently coordinate peripheral immune defense in sepsis. We now report that leptin signaling in the brain increases survival during sepsis in leptin-deficient as well as in wild-type mice and that endogenous CNS leptin action is required for an adequate systemic immune response. These findings reveal the existence of a relevant neuroendocrine control of systemic immune defense and suggest a possible therapeutic potential for leptin analogs in infectious disease.


Diabetes | 2014

Both Acyl and Des-Acyl Ghrelin Regulate Adiposity and Glucose Metabolism via Central Nervous System Ghrelin Receptors

Kristy M. Heppner; Carolin L. Piechowski; Anne Müller; Nickki Ottaway; Stephanie Sisley; David L. Smiley; Kirk M. Habegger; Paul T. Pfluger; Richard D. DiMarchi; Heike Biebermann; Matthias H. Tschöp; Darleen A. Sandoval; Diego Perez-Tilve

Growth hormone secretagogue receptors (GHSRs) in the central nervous system (CNS) mediate hyperphagia and adiposity induced by acyl ghrelin (AG). Evidence suggests that des-AG (dAG) has biological activity through GHSR-independent mechanisms. We combined in vitro and in vivo approaches to test possible GHSR-mediated biological activity of dAG. Both AG (100 nmol/L) and dAG (100 nmol/L) significantly increased inositol triphosphate formation in human embryonic kidney-293 cells transfected with human GHSR. As expected, intracerebroventricular infusion of AG in mice increased fat mass (FM), in comparison with the saline-infused controls. Intracerebroventricular dAG also increased FM at the highest dose tested (5 nmol/day). Chronic intracerebroventricular infusion of AG or dAG increased glucose-stimulated insulin secretion (GSIS). Subcutaneously infused AG regulated FM and GSIS in comparison with saline-infused control mice, whereas dAG failed to regulate these parameters even with doses that were efficacious when delivered intracerebroventricularly. Furthermore, intracerebroventricular dAG failed to regulate FM and induce hyperinsulinemia in GHSR-deficient (Ghsr−/−) mice. In addition, a hyperinsulinemic-euglycemic clamp suggests that intracerebroventricular dAG impairs glucose clearance without affecting endogenous glucose production. Together, these data demonstrate that dAG is an agonist of GHSR and regulates body adiposity and peripheral glucose metabolism through a CNS GHSR-dependent mechanism.


Journal of Clinical Investigation | 2013

p62 Links β-adrenergic input to mitochondrial function and thermogenesis

Timo D. Müller; Sang Jun Lee; Martin Jastroch; Dhiraj G. Kabra; Kerstin Stemmer; Michaela Aichler; Bill Abplanalp; Gayathri Ananthakrishnan; Nakul Bhardwaj; Sheila Collins; Senad Divanovic; Max Endele; Brian Finan; Yuanqing Gao; Kirk M. Habegger; Jazzmin Hembree; Kristy M. Heppner; Susanna M. Hofmann; Jenna Holland; Daniela Küchler; Maria Kutschke; Radha Krishna; Maarit Lehti; Rebecca Oelkrug; Nickki Ottaway; Diego Perez-Tilve; Christine Raver; Axel Walch; Sonja C. Schriever; John R. Speakman

The scaffold protein p62 (sequestosome 1; SQSTM1) is an emerging key molecular link among the metabolic, immune, and proliferative processes of the cell. Here, we report that adipocyte-specific, but not CNS-, liver-, muscle-, or myeloid-specific p62-deficient mice are obese and exhibit a decreased metabolic rate caused by impaired nonshivering thermogenesis. Our results show that p62 regulates energy metabolism via control of mitochondrial function in brown adipose tissue (BAT). Accordingly, adipocyte-specific p62 deficiency led to impaired mitochondrial function, causing BAT to become unresponsive to β-adrenergic stimuli. Ablation of p62 leads to decreased activation of p38 targets, affecting signaling molecules that control mitochondrial function, such as ATF2, CREB, PGC1α, DIO2, NRF1, CYTC, COX2, ATP5β, and UCP1. p62 ablation in HIB1B and BAT primary cells demonstrated that p62 controls thermogenesis in a cell-autonomous manner, independently of brown adipocyte development or differentiation. Together, our data identify p62 as a novel regulator of mitochondrial function and brown fat thermogenesis.

Collaboration


Dive into the Nickki Ottaway's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kirk M. Habegger

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jenna Holland

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Richard D. DiMarchi

Indiana University Bloomington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David L. Smiley

Indiana University Bloomington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge