Network


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

Hotspot


Dive into the research topics where Alexei Kharitonenkov is active.

Publication


Featured researches published by Alexei Kharitonenkov.


Trends in Endocrinology and Metabolism | 2015

FGF21 Revolutions: Recent Advances Illuminating FGF21 Biology and Medicinal Properties.

Alexei Kharitonenkov; Richard D. DiMarchi

The biology of fibroblast growth factor 21 (FGF21) has evolved through its first decade at a revolutionary pace with dramatic refinements in this relatively short span of time. This field is poised now with a deeper understanding of its specific physiological role, pathological ramifications for its inappropriate function, and a much-enriched context of the complex hormonal network in which it serves to regulate metabolism. As a derivative of these discoveries, the application of FGF21 as a medicinal agent has emerged with structurally optimized protein-based analogs being preclinically explored in multiple species, and, more recently, through clinical studies. These novel findings set a foundation for ongoing inquiries that structure future research into this intriguing protein.


Molecular metabolism | 2015

Genetic disruption of uncoupling protein 1 in mice renders brown adipose tissue a significant source of FGF21 secretion

Susanne Keipert; Maria Kutschke; Daniel Lamp; Laura Brachthäuser; Frauke Neff; Carola W. Meyer; Rebecca Oelkrug; Alexei Kharitonenkov; Martin Jastroch

Objective Circulating fibroblast growth factor 21 (FGF21) is an important auto- and endocrine player with beneficial metabolic effects on obesity and diabetes. In humans, thermogenic brown adipose tissue (BAT) was recently suggested as a source of FGF21 secretion during cold exposure. Here, we aim to clarify the role of UCP1 and ambient temperature in the regulation of FGF21 in mice. Methods Wildtype (WT) and UCP1-knockout (UCP1 KO) mice, the latter being devoid of BAT-derived non-shivering thermogenesis, were exposed to different housing temperatures. Plasma metabolites and FGF21 levels were determined, gene expression was analyzed by qPCR, and tissue histology was performed with adipose tissue. Results At thermoneutrality, FGF21 gene expression and serum levels were not different between WT and UCP1 KO mice. Cold exposure led to highly increased FGF21 serum levels in UCP1 KO mice, which were reflected in increased FGF21 gene expression in adipose tissues but not in liver and skeletal muscle. Ex vivo secretion assays revealed FGF21 release only from BAT, progressively increasing with decreasing ambient temperatures. In association with increased FGF21 serum levels in the UCP1 KO mouse, typical FGF21-related serum metabolites and inguinal white adipose tissue morphology and thermogenic gene expression were altered. Conclusions Here we show that the genetic ablation of UCP1 increases FGF21 gene expression in adipose tissue. The removal of adaptive nonshivering thermogenesis renders BAT a significant source of endogenous FGF21 under thermal stress. Thus, the thermogenic competence of BAT is not a requirement for FGF21 secretion. Notably, high endogenous FGF21 levels in UCP1-deficient models and subjects may confound pharmacological FGF21 treatments.


Molecular metabolism | 2016

Fibroblast activation protein (FAP) as a novel metabolic target

Miguel A. Sánchez-Garrido; Kirk M. Habegger; Christoffer Clemmensen; Cassie Holleman; Timo D. Müller; Diego Perez-Tilve; Pengyun Li; Archita S. Agrawal; Brian Finan; Daniel J. Drucker; Matthias H. Tschöp; Richard D. DiMarchi; Alexei Kharitonenkov

Objective Fibroblast activation protein (FAP) is a serine protease belonging to a S9B prolyl oligopeptidase subfamily. This enzyme has been implicated in cancer development and recently reported to regulate degradation of FGF21, a potent metabolic hormone. Using a known FAP inhibitor, talabostat (TB), we explored the impact of FAP inhibition on metabolic regulation in mice. Methods To address this question we evaluated the pharmacology of TB in various mouse models including those deficient in FGF21, GLP1 and GIP signaling. We also studied the ability of FAP to process FGF21 in vitro and TB to block FAP enzymatic activity. Results TB administration to diet-induced obese (DIO) animals led to profound decreases in body weight, reduced food consumption and adiposity, increased energy expenditure, improved glucose tolerance and insulin sensitivity, and lowered cholesterol levels. Total and intact plasma FGF21 were observed to be elevated in TB-treated DIO mice but not lean animals where the metabolic impact of TB was significantly attenuated. Furthermore, and in stark contrast to naïve DIO mice, the administration of TB to obese FGF21 knockout animals demonstrated no appreciable effect on body weight or any other measures of metabolism. In support of these results we observed no enzymatic degradation of human FGF21 at either end of the protein when FAP was inhibited in vitro by TB. Conclusions We conclude that pharmacological inhibition of FAP enhances levels of FGF21 in obese mice to provide robust metabolic benefits not observed in lean animals, thus validating this enzyme as a novel drug target for the treatment of obesity and diabetes.


Journal of Endocrinology | 2015

Dual effects of fibroblast growth factor 21 on hepatic energy metabolism

Ricardo J. Samms; Michelle Murphy; Maxine J Fowler; Scott Cooper; Paul J. Emmerson; Tamer Coskun; Andrew C. Adams; Alexei Kharitonenkov; Francis J. P. Ebling; Kostas Tsintzas

The aim of this study was to investigate the mechanisms by which fibroblast growth factor 21 (FGF21) affects hepatic integration of carbohydrate and fat metabolism in Siberian hamsters, a natural model of adiposity. Twelve aged matched adult male Siberian hamsters maintained in their long-day fat state since birth were randomly assigned to one of two treatment groups and were continuously infused with either vehicle (saline; n=6) or recombinant human FGF21 protein (1u200amg/kg per day; n=6) for 14 days. FGF21 administration caused a 40% suppression (P<0.05) of hepatic pyruvate dehydrogenase complex (PDC), the rate-limiting step in glucose oxidation, a 34% decrease (P<0.05) in hepatic acetylcarnitine accumulation, an index of reduced PDC flux, a 35% increase (P<0.05) in long-chain acylcarnitine content (an index of flux through β-oxidation) and a 47% reduction (P<0.05) in hepatic lipid content. These effects were underpinned by increased protein abundance of PD kinase-4 (PDK4, a negative regulator of PDC), the phosphorylated (inhibited) form of acetyl-CoA carboxylase (ACC, a negative regulator of delivery of fatty acids into the mitochondria) and the transcriptional co-regulators of energy metabolism peroxisome proliferator activated receptor gamma co-activator alpha (PGC1α) and sirtuin-1. These findings provide novel mechanistic basis to support the notion that FGF21 exerts profound metabolic benefits in the liver by modulating nutrient flux through both carbohydrate (mediated by a PDK4-mediated suppression of PDC activity) and fat (mediated by deactivation of ACC) metabolism, and therefore may be an attractive target for protection from increased hepatic lipid content and insulin resistance that frequently accompany obesity and diabetes.


Journal of Biological Chemistry | 2016

The Nuclear Receptor Rev-erbα Regulates Adipose Tissue-specific FGF21 Signaling

Jennifer Jager; Fenfen Wang; Bin Fang; Hee-Woong Lim; Lindsey C. Peed; David J. Steger; Kyoung-Jae Won; Alexei Kharitonenkov; Andrew C. Adams; Mitchell A. Lazar

FGF21 is an atypical member of the FGF family that functions as a hormone to regulate carbohydrate and lipid metabolism. Here we demonstrate that the actions of FGF21 in mouse adipose tissue, but not in liver, are modulated by the nuclear receptor Rev-erbα, a potent transcriptional repressor. Interrogation of genes induced in the absence of Rev-erbα for Rev-erbα-binding sites identified βKlotho, an essential coreceptor for FGF21, as a direct target gene of Rev-erbα in white adipose tissue but not liver. Rev-erbα ablation led to the robust elevated expression of βKlotho. Consequently, the effects of FGF21 were markedly enhanced in the white adipose tissue of mice lacking Rev-erbα. A major Rev-erbα-controlled enhancer at the Klb locus was also bound by the adipocytic transcription factor peroxisome proliferator-activated receptor (PPAR) γ, which regulates its activity in the opposite direction. These findings establish Rev-erbα as a specific modulator of FGF21 signaling in adipose tissue.


Molecular metabolism | 2018

Molecular elements in FGF19 and FGF21 defining KLB/FGFR activity and specificity

Archita S. Agrawal; Sebastian Parlee; Diego Perez-Tilve; Pengyun Li; Jia Pan; Piotr A. Mroz; Ann Maria Kruse Hansen; Birgitte Andersen; Brian Finan; Alexei Kharitonenkov; Richard D. Dimarchi

Objective To signal, FGF19 and FGF21 require co-receptor βKlotho (KLB) to act in concert with FGF receptors, and yet there is appreciable variance in the C-terminal sequences of these two novel metabolic hormones where binding is believed to be primary. We seek to determine the functional consequences for these amino acid differences and determine whether such information can be used to design high potency antagonists and agonists. Methods We employed a functional in vitro assay to identify C-terminal protein fragments capable of fully blocking KLB-mediated FGF19 and 21 receptor signaling. The key residues in each hormone responsible for support full bioactivity were identified through peptide-based Ala-scanning. Chemical optimization of the peptides was employed to increase their antagonistic potency. An optimized sequence as a substituted part of a full length FGF21 was assessed for enhanced FGFR/KLB-mediated agonism using tissue culture and obese mice. Results C-terminal FGF19 and FGF21 peptides of relatively short length were observed to potently inhibit the activity of these two hormones, in vitro and in vivo. These FGFs of different sequence also demonstrated a striking conservation of structural determinants to maintain KLB binding. A single C-terminal amino acid in FGF19 was observed to modulate relative activity through FGFR1 and FGFR4. The substitution of native FGF21 C-terminal sequence with a peptide optimized for the highest antagonistic activity resulted in significantly enhanced FGF potency, as measured by in vitro signaling and improvements in metabolic outcomes in diet-induced obese mice. Conclusions We report here the ability of short C-terminal peptides to bind KLB and function as antagonists of FGF19 and 21 actions. These proteins maintain high conservation of sequence in those residues central to KLB binding. An FGF21 chimeric protein possessing an optimized C-terminal sequence proved to be a super-agonist in delivery of beneficial metabolic effects in obese mice.


Cell Metabolism | 2014

Break on Through to the Other 1

Alexei Kharitonenkov; Richard D. DiMarchi

Although discovered in 1974, FGF1 was previously unrealized to have a role in metabolism. Suh et al. (2014) now report impressive pharmacological outcomes of FGF1 therapy in mice that imply this protein is an important metabolic regulator with therapeutic potential in the treatment of insulin resistance and type 2 diabetes.


Journal of Nutrition | 2018

Cholic Acid Supplementation of a High-Fat Obesogenic Diet Suppresses Hepatic Triacylglycerol Accumulation in Mice via a Fibroblast Growth Factor 21–Dependent Mechanism

Siri M. Ippagunta; Alexei Kharitonenkov; Andrew C. Adams; F. Bradley Hillgartner

BackgroundnSupplementation of a high-fat obesogenic diet (HFD) with cholic acid (CA) suppresses the development of obesity, insulin resistance, and hepatic steatosis in mice.nnnObjectivenWe investigated the role of fibroblast growth factor 21 (FGF21) in mediating the beneficial actions of CA on metabolic syndrome.nnnMethodsnMale 7-wk-old wild-type (WT) mice and FGF21 knockout (FGF21KO) mice were fed an HFD for 12 wk followed by a 4-wk period in which the mice were fed the HFD alone or supplemented with 0.5% CA. Body composition, gross energy efficiency, glucose tolerance, homeostasis model assessment of insulin resistance (HOMA-IR), and hepatic triacylglycerol (TG) concentrations were measured.nnnResultsnCA administration improved glucose tolerance and decreased total body fat accretion, gross energy efficiency, fasting blood glucose concentrations, and HOMA-IR in both WT mice and FGF21KO mice. The extent of the effect of CA on glucose tolerance, fasting blood glucose concentrations, and HOMA-IR was similar in both mouse strains, whereas the extent of the effect of CA on total body fat accretion and gross energy efficiency was 4.2- to 4.4-fold greater in FGF21KO mice than in WT mice. Further analyses showed that CA decreased hepatic TG concentrations in WT mice (49%) but had no effect on hepatic TG concentrations in FGF21KO mice. CA decreased the activation state of hepatic acetyl-CoA carboxylase 1 (ACC1) and adipose tissue hormone-sensitive lipase (HSL) in WT mice but was not effective in decreasing the activation of ACC1 and HSL in FGF21KO mice.nnnConclusionsnFGF21 signaling is required for the beneficial effect of CA on hepatic TG accumulation in mice fed an HFD. We propose that FGF21 signaling potentiates the ability of CA to decrease the activation of ACC1 and HSL, key enzymes controlling the supply of long-chain fatty acid precursors for hepatic TG synthesis.


Aging (Albany NY) | 2013

FGF21 drives a shift in adipokine tone to restore metabolic health

Andrew C. Adams; Alexei Kharitonenkov


Archive | 2017

Fgf21 c-terminal peptide optimization

Richard D. Dimarchi; Alexei Kharitonenkov; Pengyun Li; Archita S. Agrawal

Collaboration


Dive into the Alexei Kharitonenkov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Archita S. Agrawal

Indiana University Bloomington

View shared research outputs
Top Co-Authors

Avatar

Pengyun Li

Indiana University Bloomington

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

Amber Giles

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Bin Fang

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Bin Gao

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Cassie Holleman

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge