1
|
Huang L, Cao B, Geng Y, Zhou X, Yang Y, Ma T, Lin H, Huang Z, Zhuo L, Li J. A randomized double-blind phase Ib clinical trial of SY-009 in patients with type 2 diabetes mellitus. Eur J Pharm Sci 2024; 192:106644. [PMID: 37981049 DOI: 10.1016/j.ejps.2023.106644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 10/18/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION SY-009 produces a hypoglycemic effect via inhibiting sodium/glucose cotransporter 1 (SGLT1) in type 2 diabetes mellitus (T2DM) patients. This randomized, double-blind, placebo-controlled, and multiple-dose escalation clinical trial aimed to evaluate the pharmacokinetic and pharmacodynamical characteristics as well as the safety and tolerability of SY-009 in T2DM patients. METHOD Fifty T2DM patients were randomized into experimental and placebo groups, and hospitalized for 9 days managed with a unified diet and rest management. Subjects were given SY-009 or placebo from day 1 to day 7 at different frequencies and dosages. Single dose cohort was defined as the first dose on day 1 and multiple dose cohort included all the dose from day 1 to 7. Blood samples were collected for pharmacokinetic analysis. Mixed meal tolerance tests were performed. Blood samples were collected to determine glucose, C-peptide, insulin, glucagon-like peptide-1 (GLP-1), and gastric inhibitory polypeptide (GIP). RESULTS PK parameters were not obtained because blood SY-009 concentrations were below the limit of quantitation in all subjects. SY-009 decreased the postprandial glucose. Blood glucose was controlled within 4 hours after taking the drug. Short-term administration of SY-009 (7 days) had no significant effects on fasting glucose but reduced the secretion of C-peptide, insulin, and GIP and increased GLP-1 secretion. The most common adverse event was gastrointestinal disorder manifesting abdominal pain, diarrhea, and bloating. CONCLUSION Plasma exposure of SY-009 and its metabolites was fairly low in T2DM patients at doses of 1.0-4.0 mg. SY-009 reduced postprandial glucose, C-peptide, and insulin levels, showing relative safety and tolerability in the dose range of 1.0-4.0 mg. TRIALS REGISTRATION ClinicalTrials.gov Identifier: NCT04345107.
Collapse
Affiliation(s)
- Lei Huang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Bei Cao
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yan Geng
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiaoli Zhou
- Suzhou Yabao Pharmaceutical R&D Co., Ltd., Suzhou 215000, China
| | - Yuanxun Yang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Tingting Ma
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Hui Lin
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhijiang Huang
- Suzhou Yabao Pharmaceutical R&D Co., Ltd., Suzhou 215000, China
| | - Lang Zhuo
- Suzhou Yabao Pharmaceutical R&D Co., Ltd., Suzhou 215000, China.
| | - Juan Li
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
2
|
Liu L, Pei DS. Insights Gained from RNA Editing Targeted by the CRISPR-Cas13 Family. Int J Mol Sci 2022; 23:11400. [PMID: 36232699 PMCID: PMC9569848 DOI: 10.3390/ijms231911400] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein (Cas) systems, especially type II (Cas9) systems, have been widely developed for DNA targeting and formed a set of mature precision gene-editing systems. However, the basic research and application of the CRISPR-Cas system in RNA is still in its early stages. Recently, the discovery of the CRISPR-Cas13 type VI system has provided the possibility for the expansion of RNA targeting technology, which has broad application prospects. Most type VI Cas13 effectors have dinuclease activity that catalyzes pre-crRNA into mature crRNA and produces strong RNA cleavage activity. Cas13 can specifically recognize targeted RNA fragments to activate the Cas13/crRNA complex for collateral cleavage activity. To date, the Cas13X protein is the smallest effector of the Cas13 family, with 775 amino acids, which is a promising platform for RNA targeting due to its lack of protospacer flanking sequence (PFS) restrictions, ease of packaging, and absence of permanent damage. This study highlighted the latest progress in RNA editing targeted by the CRISPR-Cas13 family, and discussed the application of Cas13 in basic research, nucleic acid diagnosis, nucleic acid tracking, and genetic disease treatment. Furthermore, we clarified the structure of the Cas13 protein family and their molecular mechanism, and proposed a future vision of RNA editing targeted by the CRISPR-Cas13 family.
Collapse
Affiliation(s)
- Li Liu
- Chongqing Institute of Green and Intelligent Technology, Chongqing School of University of Chinese Academy of Sciences, Chinese Academy of Sciences, Chongqing 400714, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China
| | - De-Sheng Pei
- School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
3
|
Zhu Y, Li N, Huang M, Chen X, An YA, Li J, Zhao S, Funcke JB, Cao J, He Z, Zhu Q, Zhang Z, Wang ZV, Xu L, Williams KW, Li C, Grove K, Scherer PE. Activating Connexin43 gap junctions primes adipose tissue for therapeutic intervention. Acta Pharm Sin B 2022; 12:3063-3072. [PMID: 35865093 PMCID: PMC9293664 DOI: 10.1016/j.apsb.2022.02.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/16/2022] [Accepted: 02/08/2022] [Indexed: 12/19/2022] Open
Abstract
Adipose tissue is a promising target for treating obesity and metabolic diseases. However, pharmacological agents usually fail to effectively engage adipocytes due to their extraordinarily large size and insufficient vascularization, especially in obese subjects. We have previously shown that during cold exposure, connexin43 (Cx43) gap junctions are induced and activated to connect neighboring adipocytes to share limited sympathetic neuronal input amongst multiple cells. We reason the same mechanism may be leveraged to improve the efficacy of various pharmacological agents that target adipose tissue. Using an adipose tissue-specific Cx43 overexpression mouse model, we demonstrate effectiveness in connecting adipocytes to augment metabolic efficacy of the β 3-adrenergic receptor agonist Mirabegron and FGF21. Additionally, combing those molecules with the Cx43 gap junction channel activator danegaptide shows a similar enhanced efficacy. In light of these findings, we propose a model in which connecting adipocytes via Cx43 gap junction channels primes adipose tissue to pharmacological agents designed to engage it. Thus, Cx43 gap junction activators hold great potential for combination with additional agents targeting adipose tissue.
Collapse
Affiliation(s)
- Yi Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Na Li
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Mingyang Huang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi Chen
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yu A. An
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Jianping Li
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Jan-Bernd Funcke
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Jianhong Cao
- Division of Hypothalamic Research, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Zhenyan He
- Division of Hypothalamic Research, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Qingzhang Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Zhao V. Wang
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
- Department of Pediatrics, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Kevin W. Williams
- Division of Hypothalamic Research, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Chien Li
- Novo Nordisk Research Center, Seattle, WA 98109, USA
| | - Kevin Grove
- Novo Nordisk Research Center, Seattle, WA 98109, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
- Department of Cell Biology, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| |
Collapse
|
4
|
Exploring New Drug Targets for Type 2 Diabetes: Success, Challenges and Opportunities. Biomedicines 2022; 10:biomedicines10020331. [PMID: 35203540 PMCID: PMC8869656 DOI: 10.3390/biomedicines10020331] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 01/02/2023] Open
Abstract
There are substantial shortcomings in the drugs currently available for treatment of type 2 diabetes mellitus. The global diabetic crisis has not abated despite the introduction of new types of drugs and targets. Persistent unaddressed patient needs remain a significant factor in the quest for new leads in routine studies. Drug discovery methods in this area have followed developments in the market, contributing to a recent rise in the number of molecules. Nevertheless, troubling developments and fresh challenges are still evident. Recently, metformin, the most widely used first-line drug for diabetes, was found to contain a carcinogenic contaminant known as N-nitroso dimethylamine (NDMA). Therefore, purity and toxicity are also a big challenge for drug discovery and development. Moreover, newer drug classes against SGLT-2 illustrate both progress and difficulties. The same was true previously in the case of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Furthermore, researchers must study the importance of mechanistic characteristics of novel compounds, as well as exposure-related hazardous aspects of current and newly identified protein targets, in order to identify new pharmacological molecules with improved selectivity and specificity.
Collapse
|
5
|
Conceição-Furber E, Coskun T, Sloop KW, Samms RJ. Is Glucagon Receptor Activation the Thermogenic Solution for Treating Obesity? Front Endocrinol (Lausanne) 2022; 13:868037. [PMID: 35547006 PMCID: PMC9081793 DOI: 10.3389/fendo.2022.868037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/07/2022] [Indexed: 12/19/2022] Open
Abstract
A major challenge of obesity therapy is to sustain clinically relevant weight loss over time. Achieving this goal likely requires both reducing daily caloric intake and increasing caloric expenditure. Over the past decade, advances in pharmaceutical engineering of ligands targeting G protein-coupled receptors have led to the development of highly effective anorectic agents. These include mono-agonists of the GLP-1R and dual GIPR/GLP-1R co-agonists that have demonstrated substantial weight loss in experimental models and in humans. By contrast, currently, there are no medicines available that effectively augment metabolic rate to promote weight loss. Here, we present evidence indicating that activation of the GCGR may provide a solution to this unmet therapeutic need. In adult humans, GCGR agonism increases energy expenditure to a magnitude sufficient for inducing a negative energy balance. In preclinical studies, the glucagon-GCGR system affects key metabolically relevant organs (including the liver and white and brown adipose tissue) to boost whole-body thermogenic capacity and protect from obesity. Further, activation of the GCGR has been shown to augment both the magnitude and duration of weight loss that is achieved by either selective GLP-1R or dual GIPR/GLP-1R agonism in rodents. Based on the accumulation of such findings, we propose that the thermogenic activity of GCGR agonism will also complement other anti-obesity agents that lower body weight by suppressing appetite.
Collapse
|
6
|
González-Álvarez H, Bravo-Jiménez A, Martínez-Arellanes M, Gamboa-Osorio GO, Chávez-Gutiérrez E, González-Hernández LA, Gallardo-Ignacio K, Quintana-Romero OJ, Ariza-Castolo A, Guerra-Araiza C, Martino-Roaro L, Meneses-Ruiz DM, Pinto-Almazán R, Loza-Mejía MA. In Silico-Based Design and In Vivo Evaluation of an Anthranilic Acid Derivative as a Multitarget Drug in a Diet-Induced Metabolic Syndrome Model. Pharmaceuticals (Basel) 2021; 14:914. [PMID: 34577613 PMCID: PMC8466046 DOI: 10.3390/ph14090914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/13/2022] Open
Abstract
Metabolic syndrome (MetS) is a complex disease that affects almost a quarter of the world's adult population. In MetS, diabetes, obesity, hyperglycemia, high cholesterol, and high blood pressure are the most common disorders. Polypharmacy is the most used strategy for managing conditions related to MetS, but it has drawbacks such as low medication adherence. Multitarget ligands have been proposed as an interesting approach to developing drugs to treat complex diseases. However, suitable preclinical models that allow their evaluation in a context closer to a clinical situation of a complex disease are needed. From molecular docking studies, compound 1b, a 5-aminoanthranilic acid derivative substituted with 4'-trifluoromethylbenzylamino and 3',4'-dimethoxybenzamide moieties, was identified as a potential multitarget drug, as it showed high in silico affinity against targets related to MetS, including PPAR-α, PPAR-γ, and HMG-CoA reductase. It was evaluated in a diet-induced MetS rat model and simultaneously lowered blood pressure, glucose, total cholesterol, and triglyceride levels after a 14-day treatment. No toxicity events were observed during an acute lethal dose evaluation test at 1500 mg/kg. Hence, the diet-induced MetS model is suitable for evaluating treatments for MetS, and compound 1b is an attractive starting point for developing multitarget drugs.
Collapse
Affiliation(s)
- Héctor González-Álvarez
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (H.G.-Á.); (A.B.-J.); (M.M.-A.); (G.O.G.-O.); (L.M.-R.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Astrid Bravo-Jiménez
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (H.G.-Á.); (A.B.-J.); (M.M.-A.); (G.O.G.-O.); (L.M.-R.)
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Matilda Martínez-Arellanes
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (H.G.-Á.); (A.B.-J.); (M.M.-A.); (G.O.G.-O.); (L.M.-R.)
| | - Gabriela Odette Gamboa-Osorio
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (H.G.-Á.); (A.B.-J.); (M.M.-A.); (G.O.G.-O.); (L.M.-R.)
| | - Edwin Chávez-Gutiérrez
- Molecular Biology in Metabolic and Neurodegenerative Diseases Laboratory, Research Unit, High Speciality Regional Hospital of Ixtapaluca (HRAEI), Carretera Federal México-Puebla Km 34.5, Ixtapaluca 56530, Mexico; (E.C.-G.); (L.A.G.-H.); (K.G.-I.)
| | - Lino A. González-Hernández
- Molecular Biology in Metabolic and Neurodegenerative Diseases Laboratory, Research Unit, High Speciality Regional Hospital of Ixtapaluca (HRAEI), Carretera Federal México-Puebla Km 34.5, Ixtapaluca 56530, Mexico; (E.C.-G.); (L.A.G.-H.); (K.G.-I.)
| | - Karina Gallardo-Ignacio
- Molecular Biology in Metabolic and Neurodegenerative Diseases Laboratory, Research Unit, High Speciality Regional Hospital of Ixtapaluca (HRAEI), Carretera Federal México-Puebla Km 34.5, Ixtapaluca 56530, Mexico; (E.C.-G.); (L.A.G.-H.); (K.G.-I.)
| | - Osvaldo J. Quintana-Romero
- Department of Chemistry, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (O.J.Q.-R.); (A.A.-C.)
| | - Armando Ariza-Castolo
- Department of Chemistry, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (O.J.Q.-R.); (A.A.-C.)
| | - Christian Guerra-Araiza
- Medical Research Unit in Pharmacology, Specialities Hospital Bernardo Sepúlveda, National Medical Center XXI Century, Social Security Mexican Institute (IMSS), Av. Cuauhtémoc 330, Mexico City 06720, Mexico;
| | - Laura Martino-Roaro
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (H.G.-Á.); (A.B.-J.); (M.M.-A.); (G.O.G.-O.); (L.M.-R.)
- Incarnate Word University Center, Tlacoquemecatl 433, Mexico City 03100, Mexico
| | - Dulce María Meneses-Ruiz
- Noncommunicable Diseases Research Group, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico;
| | - Rodolfo Pinto-Almazán
- Molecular Biology in Metabolic and Neurodegenerative Diseases Laboratory, Research Unit, High Speciality Regional Hospital of Ixtapaluca (HRAEI), Carretera Federal México-Puebla Km 34.5, Ixtapaluca 56530, Mexico; (E.C.-G.); (L.A.G.-H.); (K.G.-I.)
| | - Marco A. Loza-Mejía
- Design, Isolation, and Synthesis of Bioactive Molecules Research Group, Chemical Sciences School, Universidad La Salle-México, Benjamín Franklin 45, Mexico City 06140, Mexico; (H.G.-Á.); (A.B.-J.); (M.M.-A.); (G.O.G.-O.); (L.M.-R.)
| |
Collapse
|
7
|
Pires Mendes C, Postal BG, Silva Frederico MJ, Gonçalves Marques Elias R, Aiceles de Medeiros Pinto V, da Fonte Ramos C, Devantier Neuenfeldt P, Nunes RJ, Mena Barreto Silva FR. Synthesis of a novel glibenclamide-pioglitazone hybrid compound and its effects on glucose homeostasis in normal and insulin-resistant rats. Bioorg Chem 2021; 114:105157. [PMID: 34328855 DOI: 10.1016/j.bioorg.2021.105157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/19/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
A new library of hybrid compounds that combine the functional parts of glibenclamide and pioglitazone was designed and developed. Compounds were screened for their antihyperglycemic effects on the glucose tolerance curve. This approach provided a single molecule that optimizes the pharmacological activities of two drugs used for the treatment of diabetes mellitus type 2 (DM2) and that have distinct biological activities, potentially minimizing the adverse effects of the original drugs. From a total of 15 compounds, 7 were evaluated in vivo; the compound 2; 4- [2- (2-phenyl-4-oxo-1,3-thiazolidin-3-yl) ethyl] benzene-1-sulfonamide (PTEBS) was selected to study its mechanism of action on glucose and lipid homeostasis in acute and chronic animal models related to DM2. PTEBS reduced glycemia and increased serum insulin in hyperglycemic rats, and elevated in vitro insulin production from isolated pancreatic islets. This compound increased the glycogen content in hepatic and muscular tissue. Moreover, PTEBS stimulated the uptake of glucose in soleus muscle through a signaling pathway similar to that of insulin, stimulating translocation and protein synthesis of glucose transporter 4 (GLUT4). PTEBS was effective in increasing insulin sensitivity in resistance rats by stimulating increased muscle glucose uptake, among other mechanisms. In addition, this compound reduced total triglycerides in a tolerance test to lipids and reduced advanced glycation end products (AGES), without altering lactate dehydrogenase (LDH) activity. Thus, we suggest that PTEBS may have similar effects to the respective prototypes, which may improve the therapeutic efficacy of these molecules and decrease adverse effects in the long-term.
Collapse
Affiliation(s)
- Camila Pires Mendes
- Universidade Federal de Santa Catarina, Departamento de Bioquímica - Centro de Ciências Biológicas, Campus Universitário, Trindade, CEP: 88040-900 - Florianópolis, SC, Brazil
| | - Bárbara Graziela Postal
- Universidade Federal de Santa Catarina, Departamento de Bioquímica - Centro de Ciências Biológicas, Campus Universitário, Trindade, CEP: 88040-900 - Florianópolis, SC, Brazil
| | - Marisa Jádna Silva Frederico
- Universidade Federal de Santa Catarina, Departamento de Bioquímica - Centro de Ciências Biológicas, Campus Universitário, Trindade, CEP: 88040-900 - Florianópolis, SC, Brazil; Universidade Federal do Ceará, Departamento de Farmacologia e Fisiologia, Faculdade de Medicina, Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Rua Coronel Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, CE 60430-275, Brazil
| | - Rui Gonçalves Marques Elias
- Universidade Federal de Santa Catarina, Departamento de Bioquímica - Centro de Ciências Biológicas, Campus Universitário, Trindade, CEP: 88040-900 - Florianópolis, SC, Brazil; Universidade Estadual do Norte do Paraná - Centro de Ciências da Saúde, Jacarezinho, PR, Brazil
| | | | | | - Patrícia Devantier Neuenfeldt
- Universidade Federal de Santa Catarina, Departamento de Química, Centro de Ciências Físicas e Matemáticas, Campus Universitário, Trindade, CEP: 88040-900, Florianópolis, SC, Brazil; Instituto Federal Catarinense, IFC, Campus São Francisco do Sul, SC, Brazil
| | - Ricardo José Nunes
- Universidade Federal de Santa Catarina, Departamento de Química, Centro de Ciências Físicas e Matemáticas, Campus Universitário, Trindade, CEP: 88040-900, Florianópolis, SC, Brazil
| | - Fátima Regina Mena Barreto Silva
- Universidade Federal de Santa Catarina, Departamento de Bioquímica - Centro de Ciências Biológicas, Campus Universitário, Trindade, CEP: 88040-900 - Florianópolis, SC, Brazil.
| |
Collapse
|
8
|
He B, Peng W, Huang J, Zhang H, Zhou Y, Yang X, Liu J, Li Z, Xu C, Xue M, Yang H, Huang P. Modulation of metabolic functions through Cas13d-mediated gene knockdown in liver. Protein Cell 2020; 11:518-524. [PMID: 32185621 PMCID: PMC7095259 DOI: 10.1007/s13238-020-00700-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Bingbing He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenbo Peng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jia Huang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hang Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yingsi Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiali Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jing Liu
- Center for Animal Genomics, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Zhijie Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunlong Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mingxing Xue
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hui Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Pengyu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
9
|
Antonellis PJ, Droz BA, Cosgrove R, O'Farrell LS, Coskun T, Perfield JW, Bauer S, Wade M, Chouinard TE, Brozinick JT, Adams AC, Samms RJ. The anti-obesity effect of FGF19 does not require UCP1-dependent thermogenesis. Mol Metab 2019; 30:131-139. [PMID: 31767164 PMCID: PMC6807368 DOI: 10.1016/j.molmet.2019.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/29/2019] [Accepted: 09/13/2019] [Indexed: 01/06/2023] Open
Abstract
Objective Fibroblast growth factor 19 (FGF19) is a postprandial hormone which plays diverse roles in the regulation of bile acid, glucose, and lipid metabolism. Administration of FGF19 to obese/diabetic mice lowers body weight, improves insulin sensitivity, and enhances glycemic control. The primary target organ of FGF19 is the liver, where it regulates bile acid homeostasis in response to nutrient absorption. In contrast, the broader pharmacologic actions of FGF19 are proposed to be driven, in part, by the recruitment of the thermogenic protein uncoupling protein 1 (UCP1) in white and brown adipose tissue. However, the precise contribution of UCP1-dependent thermogenesis to the therapeutic actions of FGF19 has not been critically evaluated. Methods Using WT and germline UCP1 knockout mice, the primary objective of the current investigation was to determine the in vivo pharmacology of FGF19, focusing on its thermogenic and anti-obesity activity. Results We report that FGF19 induced mRNA expression of UCP1 in adipose tissue and show that this effect is required for FGF19 to increase caloric expenditure. However, we demonstrate that neither UCP1 induction nor an elevation in caloric expenditure are necessary for FGF19 to induce weight loss in obese mice. In contrast, the anti-obesity action of FGF19 appeared to be associated with its known physiological role. In mice treated with FGF19, there was a significant reduction in the mRNA expression of genes associated with hepatic bile acid synthesis enzymes, lowered levels of hepatic bile acid species, and a significant increase in fecal energy content, all indicative of reduced lipid absorption in animals treated with FGF19. Conclusion Taken together, we report that the anti-obesity effect of FGF19 occurs in the absence of UCP1. Our data suggest that the primary way in which exogenous FGF19 lowers body weight in mice may be through the inhibition of bile acid synthesis and subsequently a reduction of dietary lipid absorption. FGF19 increases UCP1 transcription in adipose tissue and is associated with significant weight loss. The increase in metabolic rate observed with FGF19 is dependent upon the presence of UCP1. In the absence of UCP1, the anti-obesity effect of FGF19 are associated with a greater reduction in energy absorption.
Collapse
Affiliation(s)
- Patrick J Antonellis
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Brian A Droz
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Richard Cosgrove
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Libbey S O'Farrell
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Tamer Coskun
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - James W Perfield
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Steven Bauer
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Mark Wade
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Tara E Chouinard
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Joseph T Brozinick
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Andrew C Adams
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Ricardo J Samms
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, 46285, USA.
| |
Collapse
|
10
|
Hsieh CT, Chang FR, Tsai YH, Wu YC, Hsieh TJ. 2-Bromo-4'-methoxychalcone and 2-Iodo-4'-methoxychalcone Prevent Progression of Hyperglycemia and Obesity via 5'-Adenosine-Monophosphate-Activated Protein Kinase in Diet-Induced Obese Mice. Int J Mol Sci 2018; 19:ijms19092763. [PMID: 30223438 PMCID: PMC6163633 DOI: 10.3390/ijms19092763] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/26/2022] Open
Abstract
Obesity and diabetes are global health-threatening issues. Interestingly, the mechanism of these pathologies is quite different among individuals. The discovery and development of new categories of medicines from diverse sources are urgently needed for preventing and treating diabetes and other metabolic disorders. Previously, we reported that chalcones are important for preventing biological disorders, such as diabetes. In this study, we demonstrate that the synthetic halogen-containing chalcone derivatives 2-bromo-4′-methoxychalcone (compound 5) and 2-iodo-4′-methoxychalcone (compound 6) can promote glucose consumption and inhibit cellular lipid accumulation via 5′-adenosine-monophosphate-activated protein kinase (AMPK) activation and acetyl-CoA carboxylase 1 (ACC) phosphorylation in 3T3-L1 adipocytes and C2C12 skeletal myotubes. In addition, the two compounds significantly prevented body weight gain and impaired glucose tolerance, hyperinsulinemia, and insulin resistance, which collectively help to delay the progression of hyperglycemia in high-fat-diet-induced obese C57BL/6 mice. These findings indicate that 2-bromo-4′-methoxychalcone and 2-iodo-4′-methoxychalcone could act as AMPK activators, and may serve as lead compounds for a new class of medicines that target obesity and diabetes.
Collapse
Affiliation(s)
- Chi-Ting Hsieh
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Marine Biotechnology and Resources, College of Marine Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
| | - Yi-Hong Tsai
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yang-Chang Wu
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Tusty-Jiuan Hsieh
- Department of Marine Biotechnology and Resources, College of Marine Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
11
|
Gut P, Reischauer S, Stainier DYR, Arnaout R. LITTLE FISH, BIG DATA: ZEBRAFISH AS A MODEL FOR CARDIOVASCULAR AND METABOLIC DISEASE. Physiol Rev 2017; 97:889-938. [PMID: 28468832 PMCID: PMC5817164 DOI: 10.1152/physrev.00038.2016] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/17/2022] Open
Abstract
The burden of cardiovascular and metabolic diseases worldwide is staggering. The emergence of systems approaches in biology promises new therapies, faster and cheaper diagnostics, and personalized medicine. However, a profound understanding of pathogenic mechanisms at the cellular and molecular levels remains a fundamental requirement for discovery and therapeutics. Animal models of human disease are cornerstones of drug discovery as they allow identification of novel pharmacological targets by linking gene function with pathogenesis. The zebrafish model has been used for decades to study development and pathophysiology. More than ever, the specific strengths of the zebrafish model make it a prime partner in an age of discovery transformed by big-data approaches to genomics and disease. Zebrafish share a largely conserved physiology and anatomy with mammals. They allow a wide range of genetic manipulations, including the latest genome engineering approaches. They can be bred and studied with remarkable speed, enabling a range of large-scale phenotypic screens. Finally, zebrafish demonstrate an impressive regenerative capacity scientists hope to unlock in humans. Here, we provide a comprehensive guide on applications of zebrafish to investigate cardiovascular and metabolic diseases. We delineate advantages and limitations of zebrafish models of human disease and summarize their most significant contributions to understanding disease progression to date.
Collapse
Affiliation(s)
- Philipp Gut
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Sven Reischauer
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Didier Y R Stainier
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Rima Arnaout
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
12
|
Choi B, Lee S, Kim SM, Lee EJ, Lee SR, Kim DH, Jang JY, Kang SW, Lee KU, Chang EJ, Song JK. Dipeptidyl Peptidase-4 Induces Aortic Valve Calcification by Inhibiting Insulin-Like Growth Factor-1 Signaling in Valvular Interstitial Cells. Circulation 2017; 135:1935-1950. [DOI: 10.1161/circulationaha.116.024270] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 01/31/2017] [Indexed: 01/08/2023]
Abstract
Background:
Calcification of the aortic valve leads to increased leaflet stiffness and consequently to the development of calcific aortic valve disease. However, the underlying molecular and cellular mechanisms of calcification remain unclear. Here, we identified that dipeptidyl peptidase-4 (DPP-4, also known as CD26) increases valvular calcification and promotes calcific aortic valve disease progression.
Methods:
We obtained the aortic valve tissues from humans and murine models (wild-type and endothelial nitric oxide synthase-deficient-mice) and cultured the valvular interstitial cells (VICs) and valvular endothelial cells from the cusps. We induced osteogenic differentiation in the primary cultured VICs and examined the effects of the DPP-4 inhibitor on the osteogenic changes in vitro and aortic valve calcification in endothelial nitric oxide synthase-deficient-mice. We also induced calcific aortic stenosis in male New Zealand rabbits (weight, 2.5–3.0 kg) by a cholesterol-enriched diet+vitamin D2 (25 000 IU, daily). Echocardiography was performed to assess the aortic valve area and the maximal and mean transaortic pressure gradients at baseline and 3-week intervals thereafter. After 12 weeks, we harvested the heart and evaluated the aortic valve tissue using immunohistochemistry.
Results:
We found that nitric oxide depletion in human valvular endothelial cells activates NF-κB in human VICs. Consequently, the NF-κB promotes DPP-4 expression, which then induces the osteogenic differentiation of VICs by limiting autocrine insulin-like growth factor-1 signaling. The inhibition of DPP-4 enzymatic activity blocked the osteogenic changes in VICs in vitro and reduced the aortic valve calcification in vivo in a mouse model. Sitagliptin administration in a rabbit calcific aortic valve disease model led to significant improvements in the rate of change in aortic valve area, transaortic peak velocity, and maximal and mean pressure gradients over 12 weeks. Immunohistochemistry staining confirmed the therapeutic effect of Sitagliptin in terms of reducing the calcium deposits in the rabbit aortic valve cusps. In rabbits receiving Sitagliptin, the plasma insulin-like growth factor-1 levels were significantly increased, in line with DPP-4 inhibition.
Conclusions:
DPP-4-dependent insulin-like growth factor-1 inhibition in VICs contributes to aortic valve calcification, suggesting that DPP-4 could serve as a potential therapeutic target to inhibit calcific aortic valve disease progression.
Collapse
Affiliation(s)
- Bongkun Choi
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sahmin Lee
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Min Kim
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun-Jin Lee
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sun Ro Lee
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae-Hee Kim
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Yoon Jang
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Wook Kang
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki-Up Lee
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun-Ju Chang
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae-Kwan Song
- From Department of Biomedical Sciences (B.C., S.-M.K., E.- J.L., S.R.L., S.-W.K., E.-J.C.), Division of Cardiology (S.L., D.- H.K., J.Y.J., J.-K.S.), and Division of Endocrinology and Metabolism, Department of Internal Medicine (K.-U.L.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Lu Y, Chen C. Metabolomics: Bridging Chemistry and Biology in Drug Discovery and Development. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s40495-017-0083-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
14
|
Wang P, Liu H, Chen L, Duan Y, Chen Q, Xi S. Effects of a Novel Glucokinase Activator, HMS5552, on Glucose Metabolism in a Rat Model of Type 2 Diabetes Mellitus. J Diabetes Res 2017; 2017:5812607. [PMID: 28191470 PMCID: PMC5278194 DOI: 10.1155/2017/5812607] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/01/2016] [Accepted: 12/27/2016] [Indexed: 01/20/2023] Open
Abstract
Glucokinase (GK) plays a critical role in the control of whole-body glucose homeostasis. We investigated the possible effects of a novel glucokinase activator (GKA), HMS5552, to the GK in rats with type 2 diabetes mellitus (T2DM). Male Sprague-Dawley (SD) rats were divided into four groups: control group, diabetic group, low-dose (10 mg/kg) HMS5552-treated diabetic group (HMS-L), and high-dose (30 mg/kg) HMS5552-treated diabetic group (HMS-H). HMS5552 was administered intragastrically to the T2DM rats for one month. The levels of total cholesterol, triglyceride, fasting plasma insulin (FINS), and glucagon (FG) were determined, and an oral glucose tolerance test was performed. The expression patterns of proteins and genes associated with insulin resistance and GK activity were assayed. Compared with diabetic rats, the FINS level was significantly decreased in the HMS5552-treated diabetic rats. HMS5552 treatment significantly lowered the blood glucose levels and improved GK activity and insulin resistance. The immunohistochemistry, western blot, and semiquantitative RT-PCR results further demonstrated the effects of HMS5552 on the liver and pancreas. Our data suggest that the novel GKA, HMS5552, exerts antidiabetic effects on the liver and pancreas by improving GK activity and insulin resistance, which holds promise as a novel drug for the treatment of T2DM patients.
Collapse
Affiliation(s)
- Ping Wang
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, China
| | - Huili Liu
- School Clinic, Henan University of Science and Technology, Luoyang 471023, China
| | - Li Chen
- Department of Clinical Research & Development, Hua Medicine, Shanghai 201203, China
| | - Yingli Duan
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, China
| | - Qunli Chen
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, China
| | - Shoumin Xi
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, China
- *Shoumin Xi:
| |
Collapse
|
15
|
Ekim Üstünel B, Friedrich K, Maida A, Wang X, Krones-Herzig A, Seibert O, Sommerfeld A, Jones A, Sijmonsma TP, Sticht C, Gretz N, Fleming T, Nawroth PP, Stremmel W, Rose AJ, Berriel-Diaz M, Blüher M, Herzig S. Control of diabetic hyperglycaemia and insulin resistance through TSC22D4. Nat Commun 2016; 7:13267. [PMID: 27827363 PMCID: PMC5105165 DOI: 10.1038/ncomms13267] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/15/2016] [Indexed: 12/29/2022] Open
Abstract
Obesity-related insulin resistance represents the core component of the metabolic syndrome, promoting glucose intolerance, pancreatic beta cell failure and type 2 diabetes. Efficient and safe insulin sensitization and glucose control remain critical therapeutic aims to prevent diabetic late complications Here, we identify transforming growth factor beta-like stimulated clone (TSC) 22 D4 as a molecular determinant of insulin signalling and glucose handling. Hepatic TSC22D4 inhibition both prevents and reverses hyperglycaemia, glucose intolerance and insulin resistance in diabetes mouse models. TSC22D4 exerts its effects on systemic glucose homeostasis—at least in part—through the direct transcriptional regulation of the small secretory protein lipocalin 13 (LCN13). Human diabetic patients display elevated hepatic TSC22D4 expression, which correlates with decreased insulin sensitivity, hyperglycaemia and LCN13 serum levels. Our results establish TSC22D4 as a checkpoint in systemic glucose metabolism in both mice and humans, and propose TSC22D4 inhibition as an insulin sensitizing option in diabetes therapy. TSC22D4 regulates hepatic lipoprotein production, but has so far mainly been studied in the context of cancer cachexia. Here, the authors show TSC22D4 inhibition improves insulin sensitivity in several mouse models of diabetes, which they attribute at least in part to the induction of secreted LCN13.
Collapse
Affiliation(s)
- Bilgen Ekim Üstünel
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Kilian Friedrich
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany.,Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Adriano Maida
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Xiaoyue Wang
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Anja Krones-Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Oksana Seibert
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Anke Sommerfeld
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Allan Jones
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Tjeerd P Sijmonsma
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Carsten Sticht
- Center for Clinical Research, Medical Faculty Mannheim, 68167 Mannheim, Germany
| | - Norbert Gretz
- Center for Clinical Research, Medical Faculty Mannheim, 68167 Mannheim, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University, 69120 Heidelberg, Germany
| | - Peter P Nawroth
- Department of Medicine I and Clinical Chemistry, Heidelberg University, 69120 Heidelberg, Germany
| | - Wolfgang Stremmel
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Adam J Rose
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Mauricio Berriel-Diaz
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| |
Collapse
|
16
|
Whole-organism screening for modulators of fasting metabolism using transgenic zebrafish. Methods Mol Biol 2015; 1263:157-65. [PMID: 25618343 DOI: 10.1007/978-1-4939-2269-7_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Organismal energy homeostasis is maintained by complex interorgan communications making the discovery of novel drugs against metabolic diseases challenging using traditional high-throughput approaches in vitro. Here, we describe a method that rapidly identifies small molecules with an impact on organismal energy balance in vivo. Specifically, we developed a whole-organism screen for modulators of fasting metabolism using transgenic bioluminescence-reporter zebrafish for the gluconeogenic gene phosphoenolpyruvate-carboxykinase 1 (pck1).
Collapse
|
17
|
Chen K, Jih A, Kavaler ST, Lagakos WS, Oh D, Watkins SM, Kim JJ. Dual actions of a novel bifunctional compound to lower glucose in mice with diet-induced insulin resistance. Am J Physiol Endocrinol Metab 2015; 309:E293-301. [PMID: 26058862 PMCID: PMC4525113 DOI: 10.1152/ajpendo.00045.2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 06/04/2015] [Indexed: 01/22/2023]
Abstract
Docosahexaenoic acid (DHA 22:6n-3) and salicylate are both known to exert anti-inflammatory effects. This study investigated the effects of a novel bifunctional drug compound consisting of DHA and salicylate linked together by a small molecule that is stable in plasma but hydrolyzed in the cytoplasm. The components of the bifunctional compound acted synergistically to reduce inflammation mediated via nuclear factor κB in cultured macrophages. Notably, oral administration of the bifunctional compound acted in two distinct ways to mitigate hyperglycemia in high-fat diet-induced insulin resistance. In mice with diet-induced obesity, the compound lowered blood glucose by reducing hepatic insulin resistance. It also had an immediate glucose-lowering effect that was secondary to enhanced glucagon-like peptide-1 (GLP-1) secretion and abrogated by the administration of exendin(9-39), a GLP-1 receptor antagonist. These results suggest that the bifunctional compound could be an effective treatment for individuals with type 2 diabetes and insulin resistance. This strategy could also be employed in other disease conditions characterized by chronic inflammation.
Collapse
Affiliation(s)
- Katherine Chen
- Department of Pediatrics, University of California at San Diego, La Jolla, California
| | - Alice Jih
- Department of Pediatrics, University of California at San Diego, La Jolla, California
| | - Sarah T Kavaler
- Department of Pediatrics, University of California at San Diego, La Jolla, California
| | - William S Lagakos
- Department of Medicine, University of California at San Diego, La Jolla, California
| | - Dayoung Oh
- Department of Medicine, University of California at San Diego, La Jolla, California
| | | | - Jane J Kim
- Department of Pediatrics, University of California at San Diego, La Jolla, California; Rady Children's Hospital of San Diego, San Diego, California
| |
Collapse
|
18
|
Stegemann R, Buchner DA. Transgenerational inheritance of metabolic disease. Semin Cell Dev Biol 2015; 43:131-140. [PMID: 25937492 PMCID: PMC4626440 DOI: 10.1016/j.semcdb.2015.04.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 04/20/2015] [Accepted: 04/22/2015] [Indexed: 02/05/2023]
Abstract
Metabolic disease encompasses several disorders including obesity, type 2 diabetes, and dyslipidemia. Recently, the incidence of metabolic disease has drastically increased, driven primarily by a worldwide obesity epidemic. Transgenerational inheritance remains controversial, but has been proposed to contribute to human metabolic disease risk based on a growing number of proof-of-principle studies in model organisms ranging from Caenorhabditis elegans to Mus musculus to Sus scrofa. Collectively, these studies demonstrate that heritable risk is epigenetically transmitted from parent to offspring over multiple generations in the absence of a continued exposure to the triggering stimuli. A diverse assortment of initial triggers can induce transgenerational inheritance including high-fat or high-sugar diets, low-protein diets, various toxins, and ancestral genetic variants. Although the mechanistic basis underlying the transgenerational inheritance of disease risk remains largely unknown, putative molecules mediating transmission include small RNAs, histone modifications, and DNA methylation. Due to the considerable impact of metabolic disease on human health, it is critical to better understand the role of transgenerational inheritance of metabolic disease risk to open new avenues for therapeutic intervention and improve upon the current methods for clinical diagnoses and treatment.
Collapse
Affiliation(s)
- Rachel Stegemann
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States; Department of Biological Chemistry, Case Western Reserve University, Cleveland, OH 44106, United States.
| |
Collapse
|
19
|
Samms RJ, Smith DP, Cheng CC, Antonellis PP, Perfield JW, Kharitonenkov A, Gimeno RE, Adams AC. Discrete Aspects of FGF21 In Vivo Pharmacology Do Not Require UCP1. Cell Rep 2015; 11:991-9. [PMID: 25956583 DOI: 10.1016/j.celrep.2015.04.046] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/12/2015] [Accepted: 04/22/2015] [Indexed: 12/30/2022] Open
Abstract
A primary target of the pleiotropic metabolic hormone FGF21 is adipose tissue, where it initiates a gene expression program to enhance energy expenditure, an effect presumed to be centered on augmented UCP1 expression and activity. In UCP1 null (UCP1KO) mice, we show that the effect of FGF21 to increase the metabolic rate is abolished. However, in contrast to prior expectations, we found that increased UCP1-dependent thermogenesis is only partially required to achieve the beneficial effects of FGF21 treatment. In UCP1KO mice, there appears to be an underlying reduction in food intake following FGF21 administration, facilitating weight loss equal to that observed in wild-type animals. Furthermore, we show that UCP1-dependent thermogenesis is not required for FGF21 to improve glycemic control or to reduce circulating cholesterol or free fatty acids. These data indicate that several important metabolic endpoints of FGF21 are UCP1 independent; however, the contribution of UCP1-dependent thermogenesis to other discrete aspects of FGF21 biology requires further study.
Collapse
Affiliation(s)
- Ricardo J Samms
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Dennis P Smith
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Christine C Cheng
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Patrick P Antonellis
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - James W Perfield
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Alexei Kharitonenkov
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Ruth E Gimeno
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Andrew C Adams
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| |
Collapse
|
20
|
Yen HF, Hsieh CT, Hsieh TJ, Chang FR, Wang CK. In vitro anti-diabetic effect and chemical component analysis of 29 essential oils products. J Food Drug Anal 2015; 23:124-129. [PMID: 28911435 PMCID: PMC9351752 DOI: 10.1016/j.jfda.2014.02.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/22/2014] [Accepted: 02/26/2014] [Indexed: 01/05/2023] Open
|
21
|
Lu M, Li P, Bandyopadhyay G, Lagakos W, DeWolf WE, Alford T, Chicarelli MJ, Williams L, Anderson DA, Baer BR, McVean M, Conn M, Véniant MM, Coward P. Characterization of a novel glucokinase activator in rat and mouse models. PLoS One 2014; 9:e88431. [PMID: 24533087 PMCID: PMC3922816 DOI: 10.1371/journal.pone.0088431] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 01/07/2014] [Indexed: 12/14/2022] Open
Abstract
Glucokinase (GK) is a hexokinase isozyme that catalyzes the phosphorylation of glucose to glucose-6-phosphate. Glucokinase activators are being investigated as potential diabetes therapies because of their effects on hepatic glucose output and/or insulin secretion. Here, we have examined the efficacy and mechanisms of action of a novel glucokinase activator, GKA23. In vitro, GKA23 increased the affinity of rat and mouse glucokinase for glucose, and increased glucose uptake in primary rat hepatocytes. In vivo, GKA23 treatment improved glucose homeostasis in rats by enhancing beta cell insulin secretion and suppressing hepatic glucose production. Sub-chronic GKA23 treatment of mice fed a high-fat diet resulted in improved glucose homeostasis and lipid profile.
Collapse
Affiliation(s)
- Min Lu
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Pingping Li
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Gautam Bandyopadhyay
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - William Lagakos
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Walter E. DeWolf
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | - Taylor Alford
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | | | - Lance Williams
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | | | - Brian R. Baer
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | - Maralee McVean
- Array BioPharma Inc., Boulder, Colorado, United States of America
| | - Marion Conn
- Amgen Inc., South San Francisco, California, United States of America
| | | | - Peter Coward
- Amgen Inc., South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
22
|
Kharitonenkov A, Adams AC. Inventing new medicines: The FGF21 story. Mol Metab 2013; 3:221-9. [PMID: 24749049 PMCID: PMC3986619 DOI: 10.1016/j.molmet.2013.12.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 12/17/2013] [Accepted: 12/19/2013] [Indexed: 12/20/2022] Open
Abstract
Since the discovery of insulin in 1921, protein therapeutics have become vital tools in the treatment of diabetes mellitus. This heritage has been extended with the comparatively recent introduction of recombinant and re-engineered insulins, in addition to the advent of GLP1 agonists. FGF21 represents an example of a novel experimental protein therapy which is able to induce favorable metabolic effects in various species ranging from rodents to man. The aim of this review is to communicate the story of the FGF21 drug discovery path from identification in a functional in vitro screen, to the eventual evaluation of its utility in patients. Given that the development of FGF21 advanced hand-in-hand with rapidly evolving scientific research around this target, we have also attempted to describe our view of recent developments regarding the mechanistic understanding of FGF21 biology.
Collapse
|
23
|
Gaich G, Chien JY, Fu H, Glass LC, Deeg MA, Holland WL, Kharitonenkov A, Bumol T, Schilske HK, Moller DE. The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes. Cell Metab 2013; 18:333-40. [PMID: 24011069 DOI: 10.1016/j.cmet.2013.08.005] [Citation(s) in RCA: 682] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 07/02/2013] [Accepted: 07/24/2013] [Indexed: 01/13/2023]
Abstract
Fibroblast growth factor 21 (FGF21) is a recently discovered metabolic regulator. Exogenous FGF21 produces beneficial metabolic effects in animal models; however, the translation of these observations to humans has not been tested. Here, we studied the effects of LY2405319 (LY), a variant of FGF21, in a randomized, placebo-controlled, double-blind proof-of-concept trial in patients with obesity and type 2 diabetes. Patients received placebo or 3, 10, or 20 mg of LY daily for 28 days. LY treatment produced significant improvements in dyslipidemia, including decreases in low-density lipoprotein cholesterol and triglycerides and increases in high-density lipoprotein cholesterol and a shift to a potentially less atherogenic apolipoprotein concentration profile. Favorable effects on body weight, fasting insulin, and adiponectin were also detected. However, only a trend toward glucose lowering was observed. These results indicate that FGF21 is bioactive in humans and suggest that FGF21-based therapies may be effective for the treatment of selected metabolic disorders.
Collapse
Affiliation(s)
- Gregory Gaich
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
He S, Hong Q, Lai Z, Wu Z, Yu Y, Kim DW, Ting PC, Kuethe JT, Yang GX, Jian T, Liu J, Guiadeen D, Krikorian AD, Sperbeck DM, Sonatore LM, Wiltsie J, Chung CC, Gibson JT, Lisnock J, Murphy BA, Gorski JN, Liu J, Chen D, Chen X, Wolff M, Tong SX, Madeira M, Karanam BV, Shen DM, Balkovec JM, Pinto S, Nargund RP, DeVita RJ. Potent DGAT1 Inhibitors in the Benzimidazole Class with a Pyridyl-oxy-cyclohexanecarboxylic Acid Moiety. ACS Med Chem Lett 2013; 4:773-8. [PMID: 24900745 DOI: 10.1021/ml400168h] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 06/06/2013] [Indexed: 12/24/2022] Open
Abstract
We report the design and synthesis of a series of novel DGAT1 inhibitors in the benzimidazole class with a pyridyl-oxy-cyclohexanecarboxylic acid moiety. In particular, compound 11A is a potent DGAT1 inhibitor with excellent selectivity against ACAT1. Compound 11A significantly reduces triglyceride excursion in lipid tolerance tests (LTT) in both mice and dogs at low plasma exposure. An in vivo study in mice with des-fluoro analogue 10A indicates that this series of compounds appears to distribute in intestine preferentially over plasma. The propensity to target intestine over plasma could be advantageous in reducing potential side effects since lower circulating levels of drug are required for efficacy. However, in the preclinical species, compound 11A undergoes cis/trans epimerization in vivo, which could complicate further development due to the presence of an active metabolite.
Collapse
Affiliation(s)
- Shuwen He
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Qingmei Hong
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Zhong Lai
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Zhicai Wu
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Yang Yu
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - David W. Kim
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Pauline C. Ting
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Jeffrey T. Kuethe
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Ginger X. Yang
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Tianying Jian
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Jian Liu
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Deodial Guiadeen
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Arto D. Krikorian
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Donald M. Sperbeck
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Lisa M. Sonatore
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Judyann Wiltsie
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Christine C. Chung
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Jack T. Gibson
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - JeanMarie Lisnock
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Beth A. Murphy
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Judith N. Gorski
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Jinqi Liu
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Dunlu Chen
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Xiaoli Chen
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Michael Wolff
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Sharon X. Tong
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Maria Madeira
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Bindhu V. Karanam
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Dong-Ming Shen
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - James M. Balkovec
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Shirly Pinto
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Ravi P. Nargund
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| | - Robert J. DeVita
- Discovery and Preclinical Sciences, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth,
New Jersey 07033, United States
| |
Collapse
|
25
|
Adams AC, Halstead CA, Hansen BC, Irizarry AR, Martin JA, Myers SR, Reynolds VL, Smith HW, Wroblewski VJ, Kharitonenkov A. LY2405319, an Engineered FGF21 Variant, Improves the Metabolic Status of Diabetic Monkeys. PLoS One 2013; 8:e65763. [PMID: 23823755 PMCID: PMC3688819 DOI: 10.1371/journal.pone.0065763] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/29/2013] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a novel metabolic regulator that represents a promising target for the treatment of several metabolic diseases. Administration of recombinant wild type FGF21 to diabetic animals leads to a dramatic improvement in glycaemia and ameliorates other systemic measures of metabolic health. Here we report the pharmacologic outcomes observed in non-human primates upon administration of a recently described FGF21 analogue, LY2405319 (LY). Diabetic rhesus monkeys were treated subcutaneously with LY once daily for a period of seven weeks. The doses of LY used were 3, 9 and 50 mg/kg each delivered in an escalating fashion with washout measurements taken at 2, 4, 6 and 8 weeks following the final LY dose. LY therapy led to a dramatic and rapid lowering of several important metabolic parameters including glucose, body weight, insulin, cholesterol and triglyceride levels at all doses tested. In addition, we observed favorable changes in circulating profiles of adipokines, with increased adiponectin and reduced leptin indicative of direct FGF21 action on adipose tissue. Importantly, and for the first time we show that FGF21 based therapy has metabolic efficacy in an animal with late stage diabetes. While the glycemic efficacy of LY in this animal was partially attenuated its lipid lowering effect was fully preserved suggesting that FGF21 may be a viable treatment option even in patients with advanced disease progression. These findings support continued exploration of the FGF21 pathway for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Andrew C. Adams
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Carolyn A. Halstead
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Barbara C. Hansen
- Center for Preclinical Research, University of South Florida, Tampa, Florida, United States of America
| | - Armando R. Irizarry
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Jennifer A. Martin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Sharon R. Myers
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Vincent L. Reynolds
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Holly W. Smith
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Victor J. Wroblewski
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Alexei Kharitonenkov
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
26
|
Rational design of a fibroblast growth factor 21-based clinical candidate, LY2405319. PLoS One 2013; 8:e58575. [PMID: 23536797 PMCID: PMC3594191 DOI: 10.1371/journal.pone.0058575] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 02/05/2013] [Indexed: 01/08/2023] Open
Abstract
Fibroblast growth factor 21 is a novel hormonal regulator with the potential to treat a broad variety of metabolic abnormalities, such as type 2 diabetes, obesity, hepatic steatosis, and cardiovascular disease. Human recombinant wild type FGF21 (FGF21) has been shown to ameliorate metabolic disorders in rodents and non-human primates. However, development of FGF21 as a drug is challenging and requires re-engineering of its amino acid sequence to improve protein expression and formulation stability. Here we report the design and characterization of a novel FGF21 variant, LY2405319. To enable the development of a potential drug product with a once-daily dosing profile, in a preserved, multi-use formulation, an additional disulfide bond was introduced in FGF21 through Leu118Cys and Ala134Cys mutations. FGF21 was further optimized by deleting the four N-terminal amino acids, His-Pro-Ile-Pro (HPIP), which was subject to proteolytic cleavage. In addition, to eliminate an O-linked glycosylation site in yeast a Ser167Ala mutation was introduced, thus allowing large-scale, homogenous protein production in Pichia pastoris. Altogether re-engineering of FGF21 led to significant improvements in its biopharmaceutical properties. The impact of these changes was assessed in a panel of in vitro and in vivo assays, which confirmed that biological properties of LY2405319 were essentially identical to FGF21. Specifically, subcutaneous administration of LY2405319 in ob/ob and diet-induced obese (DIO) mice over 7–14 days resulted in a 25–50% lowering of plasma glucose coupled with a 10–30% reduction in body weight. Thus, LY2405319 exhibited all the biopharmaceutical and biological properties required for initiation of a clinical program designed to test the hypothesis that administration of exogenous FGF21 would result in effects on disease-related metabolic parameters in humans.
Collapse
|
27
|
|
28
|
|
29
|
Coppari R, Bjørbæk C. Leptin revisited: its mechanism of action and potential for treating diabetes. Nat Rev Drug Discov 2012; 11:692-708. [PMID: 22935803 PMCID: PMC4019022 DOI: 10.1038/nrd3757] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the discovery of leptin in 1994, we now have a better understanding of the cellular and molecular mechanisms underlying its biological effects. In addition to its established anti-obesity effects, leptin exerts antidiabetic actions that are independent of its regulation of body weight and food intake. In particular, leptin can correct diabetes in animal models of type 1 and type 2 diabetes. In addition, long-term leptin replacement therapy improves glycaemic control, insulin sensitivity and plasma triglycerides in patients with severe insulin resistance due to lipodystrophy. These results have spurred enthusiasm for the use of leptin therapy to treat diabetes. Here, we review the current understanding of the glucoregulatory functions of leptin, emphasizing its central mechanisms of action and lessons learned from clinical studies, and discuss possible therapeutic applications of leptin in the treatment of type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Roberto Coppari
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas TX, 75390, USA
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- The Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, 92697, USA
| | - Christian Bjørbæk
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston MA, 02215, USA
| |
Collapse
|
30
|
Gilliam LAA, Neufer PD. Transgenic mouse models resistant to diet-induced metabolic disease: is energy balance the key? J Pharmacol Exp Ther 2012; 342:631-6. [PMID: 22700428 DOI: 10.1124/jpet.112.192146] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The prevalence and economic burden of obesity and type 2 diabetes is a driving force for the discovery of molecular targets to improve insulin sensitivity and glycemic control. Here, we review several transgenic mouse models that identify promising targets, ranging from proteins involved in the insulin signaling pathway, alterations of genes affecting energy metabolism, and transcriptional metabolic regulators. Despite the diverse endpoints in each model, a common thread that emerges is the necessity for maintenance of energy balance, suggesting pharmacotherapy must target the development of drugs that decrease energy intake, accelerate energy expenditure in a well controlled manner, or augment natural compensatory responses to positive energy balance.
Collapse
Affiliation(s)
- Laura A A Gilliam
- Department of Physiology, East Carolina University, Greenville, NC, USA
| | | |
Collapse
|
31
|
Piceatannol, a resveratrol derivative, promotes glucose uptake through glucose transporter 4 translocation to plasma membrane in L6 myocytes and suppresses blood glucose levels in type 2 diabetic model db/db mice. Biochem Biophys Res Commun 2012; 422:469-75. [DOI: 10.1016/j.bbrc.2012.05.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 05/03/2012] [Indexed: 01/27/2023]
|