Network


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

Hotspot


Dive into the research topics where Vasily Gelfanov is active.

Publication


Featured researches published by Vasily Gelfanov.


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.


Oncogene | 2000

Paclitaxel sensitivity of breast cancer cells with constitutively active NF-κB is enhanced by IκBα super-repressor and parthenolide

Nikhil M. Patel; Shinichi Nozaki; Nicholas H. Shortle; Poornima Bhat-Nakshatri; Thomas R. Newton; Susan Rice; Vasily Gelfanov; Scott H. Boswell; Robert J. Goulet; George W. Sledge; Harikrishna Nakshatri

The transcription factor nuclear factor-κB (NF-κB) regulates genes important for tumor invasion, metastasis and chemoresistance. Normally, NF-κB remains sequestered in an inactive state by cytoplasmic inhibitor-of-κB (IκB) proteins. NF-κB translocates to nucleus and activates gene expression upon exposure of cells to growth factors and cytokines. We and others have shown previously that NF-κB is constitutively active in a subset of breast cancers. In this study, we show that constitutive activation of NF-κB leads to overexpression of the anti-apoptotic genes c-inhibitor of apoptosis 2 (c-IAP2) and manganese superoxide dismutase (Mn-SOD) in breast cancer cells. Furthermore, expression of the anti-apoptotic tumor necrosis factor receptor associated factor 1 (TRAF1) and defender-against cell death (DAD-1) is regulated by NF-κB in certain breast cancer cells. We also demonstrate that NF-κB-inducible genes protect cancer cells against paclitaxel as MDA-MB-231 breast cancer cells modified to overexpress IκBα required lower concentrations of paclitaxel to arrest at the G2/M phase of the cell cycle and undergo apoptosis when compared to parental cells. The effect of NF-κB on paclitaxel-sensitivity appears to be specific to cancer cells because normal fibroblasts derived from embryos lacking p65 subunit of NF-κB and wild type littermate embryos were insensitive to paclitaxel-induced G2/M cell cycle arrest. Parthenolide, an active ingredient of herbal remedies such as feverfew (tanacetum parthenium), mimicked the effects of IκBα by inhibiting NF-κB DNA binding activity and Mn-SOD expression, and increasing paclitaxel-induced apoptosis of breast cancer cells. These results suggest that active ingredients of herbs with anti-inflammatory properties may be useful in increasing the sensitivity of cancers with constitutively active NF-κB to chemotherapeutic drugs.


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.


Physiology & Behavior | 2010

Glucagon regulation of energy metabolism.

Kristy M. Heppner; Kirk M. Habegger; Jonathan Day; Paul T. Pfluger; Diego Perez-Tilve; Brian P. Ward; Vasily Gelfanov; Steve Woods; Richard D. DiMarchi; Matthias H. Tschöp

Glucagon has long been known as a counter-regulatory hormone to insulin of fundamental importance to glucose homeostasis. Its prominent ability to stimulate glycogenolysis and gluconeogenesis, has historically cast this peptide as one hormone where the metabolic consequences of increasing blood glucose levels, especially in obesity, are viewed largely as being deleterious. This perspective may be changing in light of emerging data and reconsideration of historic studies, which suggest that glucagon has beneficial effects on body fat mass, food intake, and energy expenditure. In this review, we discuss the mechanisms of glucagon-mediated body weight regulation as well as possible novel therapeutic approaches in the treatment of obesity and glucose intolerance that may arise from these findings. The paper represents an invited review by a symposium, award winner or keynote speaker at the Society for the Study of Ingestive Behavior [SSIB] Annual Meeting in Portland, July 2009.


Cell Metabolism | 2016

Unimolecular Polypharmacy for Treatment of Diabetes and Obesity

Matthias H. Tschöp; Brian Finan; Christoffer Clemmensen; Vasily Gelfanov; Diego Perez-Tilve; Timo D. Müller; Richard D. DiMarchi

Many complex diseases have historically proven to be defiant to the best mono-therapeutic approaches. Several examples of combination therapies have largely overcome such challenges, notably for the treatment of severe hypertension and tuberculosis. Obesity and its consequences, such as type 2 diabetes, have proven to be equally resistant to therapeutic approaches based on single medicines. Proper management of type 2 diabetes often requires adjunctive medications, and the recent registration of a few compound mixtures has set the precedent for combinatorial treatment of obesity. On the other hand, double or triple therapeutic combinations are more difficult to advance to regulatory approval than single molecules. More recently, several classes of novel unimolecular combination therapeutics have emerged with superior efficacy than currently prescribed options and pose the potential to reverse obesity and type 2 diabetes. Here, we summarize the discovery, pre-clinical validation, and first clinical test of such peptide hormone poly-agonist drug candidates.


Biopolymers | 2012

Optimization of co‐agonism at GLP‐1 and glucagon receptors to safely maximize weight reduction in DIO‐rodents

Jonathan Day; Vasily Gelfanov; David L. Smiley; Paul E. Carrington; George J. Eiermann; Gary G. Chicchi; Mark D. Erion; Jas Gidda; Nancy A. Thornberry; Matthias H. Tschöp; Donald J. Marsh; Ranabir SinhaRoy; Richard D. DiMarchi; Alessandro Pocai

The ratio of GLP‐1/glucagon receptor (GLP1R/GCGR) co‐agonism that achieves maximal weight loss without evidence of hyperglycemia was determined in diet‐induced obese (DIO) mice chronically treated with GLP1R/GCGR co‐agonist peptides differing in their relative receptor agonism. Using glucagon‐based peptides, a spectrum of receptor selectivity was achieved by a combination of selective incorporation of GLP‐1 sequences, C‐terminal modification, backbone lactam stapling to stabilize helical structure, and unnatural amino acid substitutions at the N‐terminal dipeptide. In addition to α‐amino‐isobutyric acid (Aib) substitution at position two, we show that α,α′‐dimethyl imidazole acetic acid (Dmia) can serve as a potent replacement for the highly conserved histidine at position one. Selective site‐specific pegylation was used to further minimize enzymatic degradation and provide uniform, extended in vivo duration of action. Maximal weight loss devoid of any sign of hyperglycemia was achieved with a co‐agonist comparably balanced for in vitro potency at murine GLP1R and GCGR. This peptide exhibited superior weight loss and glucose lowering compared to a structurally matched pure GLP1R agonist, and to co‐agonists of relatively reduced GCGR tone. Any further enhancement of the relative GCGR agonist potency yielded increased weight loss but at the expense of elevated blood glucose. We conclude that GCGR agonism concomitant with GLP1R agonism constitutes a promising approach to treatment of the metabolic syndrome. However, the relative ratio of GLP1R/GCGR co‐agonism needs to be carefully chosen for each species to maximize weight loss efficacy and minimize hyperglycemia.


Cell | 2016

Chemical Hybridization of Glucagon and Thyroid Hormone Optimizes Therapeutic Impact for Metabolic Disease.

Brian Finan; Christoffer Clemmensen; Zhimeng Zhu; Kerstin Stemmer; Karine Gauthier; Luisa Müller; Meri De Angelis; Kristin Moreth; Frauke Neff; Diego Perez-Tilve; Katrin Fischer; Dominik Lutter; Miguel A. Sánchez-Garrido; Peng Liu; Jan Tuckermann; Mohsen Malehmir; Marc E. Healy; Achim Weber; Mathias Heikenwalder; Martin Jastroch; Maximilian Kleinert; Sigrid Jall; Sara J. Brandt; Frédéric Flamant; Karl-Werner Schramm; Heike Biebermann; Yvonne Döring; Christian Weber; Kirk M. Habegger; Michaela Keuper

Glucagon and thyroid hormone (T3) exhibit therapeutic potential for metabolic disease but also exhibit undesired effects. We achieved synergistic effects of these two hormones and mitigation of their adverse effects by engineering chemical conjugates enabling delivery of both activities within one precisely targeted molecule. Coordinated glucagon and T3 actions synergize to correct hyperlipidemia, steatohepatitis, atherosclerosis, glucose intolerance, and obesity in metabolically compromised mice. We demonstrate that each hormonal constituent mutually enriches cellular processes in hepatocytes and adipocytes via enhanced hepatic cholesterol metabolism and white fat browning. Synchronized signaling driven by glucagon and T3 reciprocally minimizes the inherent harmful effects of each hormone. Liver-directed T3 action offsets the diabetogenic liability of glucagon, and glucagon-mediated delivery spares the cardiovascular system from adverse T3 action. Our findings support the therapeutic utility of integrating these hormones into a single molecular entity that offers unique potential for treatment of obesity, type 2 diabetes, and cardiovascular disease.

Collaboration


Dive into the Vasily Gelfanov's collaboration.

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

Nickki Ottaway

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jonathan Day

Indiana University Bloomington

View shared research outputs
Top Co-Authors

Avatar

Kirk M. Habegger

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

James T. Patterson

Indiana University Bloomington

View shared research outputs
Top Co-Authors

Avatar

Bin Yang

Indiana University Bloomington

View shared research outputs
Researchain Logo
Decentralizing Knowledge