1
|
Uehara Y, Komatsu T, Sasaki K, Abe S, Nakashima S, Yamamoto T, Kim JE, Cho KH. Cuban policosanol improves high-density lipoprotein cholesterol efflux capacity in healthy Japanese subjects. Front Nutr 2024; 10:1297008. [PMID: 38260075 PMCID: PMC10800607 DOI: 10.3389/fnut.2023.1297008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Policosanol supplementation has been reported to increase high-density lipoprotein (HDL)-cholesterol (HDL-C). However, the association between Cuban policosanol supplementation and HDL cholesterol efflux capacity (CEC), an important function of HDL, remains unclear. We performed a lipoprotein analysis investigating 32 Japanese healthy participants (placebo, n = 17 or policosanol supplementation for 12 weeks, n = 15) from a randomized Cuban policosanol clinical trial. First, HDL CEC and HDL-related factors were measured before and after policosanol supplementation. Then, through electron microscopy after ultracentrifugation and high-performance liquid chromatography, HDL morphology and subclass were analyzed, respectively. Finally, the effects of policosanol supplementation regarding HDL function, HDL-related factors, and HDL morphology/component were examined. Cuban policosanol considerably increased the HDL CEC and HDL-C and apolipoprotein A-I (ApoA-I) levels. Furthermore, policosanol supplementation led to larger HDL particles, increased cholesterol content in larger HDL particles, and reduced triglyceride content in smaller HDL particles. In participants with high baseline HDL-C levels, the policosanol effects for HDL CEC are observed. HDL CEC fluctuation induced by policosanol was highly associated with HDL-C and ApoA-I changes. In conclusion, for the first time, we demonstrated that policosanol supplementation increased the HDL CEC in healthy participants.
Collapse
Affiliation(s)
- Yoshinari Uehara
- Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
- Research Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Center for Preventive, Anti-aging and Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Tomohiro Komatsu
- Research Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Center for Preventive, Anti-aging and Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Kei Sasaki
- Center for Preventive, Anti-aging and Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Satomi Abe
- Research Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
| | - Shihoko Nakashima
- Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Taiki Yamamoto
- Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Ji-Eun Kim
- Raydel Research Institute, Medical Innovation Complex, Daegu, Republic of Korea
| | - Kyung-Hyun Cho
- Raydel Research Institute, Medical Innovation Complex, Daegu, Republic of Korea
- LipoLab, Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
2
|
Chang CK, Chiang EPI, Chang KH, Tang KT, Chen PK, Yip HT, Chen CH, Chen DY. The Sizes and Composition of HDL-Cholesterol Are Significantly Associated with Inflammation in Rheumatoid Arthritis Patients. Int J Mol Sci 2023; 24:10645. [PMID: 37445823 DOI: 10.3390/ijms241310645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Rheumatoid arthritis (RA), a chronic inflammatory disease, carries a significant burden of atherosclerotic cardiovascular diseases (ASCVD). With their heterogeneous composition, high-density lipoprotein (HDL) particles have varied athero-protective properties, and some may even increase ASCVD risk. In this prospective and cross-sectional study, we aimed to examine the relationship between HDL sizes/metabolites and inflammation in RA. Using 1H-NMR-based lipid/metabolomics, differential HDL-related metabolites were identified between RA patients and healthy control (HC) subjects and between RA patients with and without anti-citrullinated peptide antibodies (ACPA). The correlation between the discriminative HDL-related metabolites and C-reactive protein (CRP) was evaluated in RA patients. RA patients demonstrated higher particle number, lipids, cholesterol, cholesterol ester, free cholesterol, and phospholipids in large/very large-sized HDLs. ACPA-positive patients had higher L-HDL-C and L-HDL-CE but lower small-/medium-sized HDL-TG levels than ACPA-negative patients. An inverse correlation was found between CRP levels and small-sized HDLs. Janus kinase inhibitor treatment was associated with increased serum small-sized HDL-related metabolites and decreased CRP levels. We are the first to reveal the significant associations between RA inflammation and HDL sizes/metabolites. A potential link between ACPA positivity and changes in serum levels of HDL-related metabolites was also observed in RA patients.
Collapse
Affiliation(s)
- Ching-Kun Chang
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung 404, Taiwan
- Translational Medicine Laboratory, Rheumatology Research Center, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| | - En-Pei Isabel Chiang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Innovation and Development Center of Sustainable Agriculture, National Chung Hsing University, Taichung 402, Taiwan
| | - Kuang-Hsi Chang
- Department of Medical Research, Tungs' Taichung Metroharbor Hospital, Taichung 435, Taiwan
- Center for General Education, China Medical University, Taichung 404, Taiwan
- General Education Center, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
| | - Kuo-Tung Tang
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Po-Ku Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung 404, Taiwan
- Translational Medicine Laboratory, Rheumatology Research Center, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Hei-Tung Yip
- College of Medicine, China Medical University, Taichung 404, Taiwan
- Management Office for Health Data, China Medical University Hospital, Taichung 404, Taiwan
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA
- Institute for Biomedical Sciences, Shinshu University, Nagano 390-8621, Japan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung 404, Taiwan
- Translational Medicine Laboratory, Rheumatology Research Center, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
- College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
3
|
Satapati S, Downes DP, Metzger D, Shankaran H, Talukdar S, Zhou Y, Ren Z, Chen M, Lim YH, Hatcher NG, Wen X, Sheth PR, McLaren DG, Previs SF. Using measures of metabolic flux to align screening and clinical development: Avoiding pitfalls to enable translational studies. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:20-28. [PMID: 35058172 DOI: 10.1016/j.slasd.2021.10.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Screening campaigns, especially those aimed at modulating enzyme activity, often rely on measuring substrate→product conversions. Unfortunately, the presence of endogenous substrates and/or products can limit one's ability to measure conversions. As well, coupled detection systems, often used to facilitate optical readouts, are subject to interference. Stable isotope labeled substrates can overcome background contamination and yield a direct readout of enzyme activity. Not only can isotope kinetic assays enable early screening, but they can also be used to follow hit progression in translational (pre)clinical studies. Herein, we consider a case study surrounding lipid biology to exemplify how metabolic flux analyses can connect stages of drug development, caveats are highlighted to ensure reliable data interpretations. For example, when measuring enzyme activity in early biochemical screening it may be enough to quantify the formation of a labeled product. In contrast, cell-based and in vivo studies must account for variable exposure to a labeled substrate (or precursor) which occurs via tracer dilution and/or isotopic exchange. Strategies are discussed to correct for these complications. We believe that measures of metabolic flux can help connect structure-activity relationships with pharmacodynamic mechanisms of action and determine whether mechanistically differentiated biophysical interactions lead to physiologically relevant outcomes. Adoption of this logic may allow research programs to (i) build a critical bridge between primary screening and (pre)clinical development, (ii) elucidate biology in parallel with screening and (iii) suggest a strategy aimed at in vivo biomarker development.
Collapse
Affiliation(s)
- Santhosh Satapati
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Daniel P Downes
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Daniel Metzger
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Harish Shankaran
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Saswata Talukdar
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Yingjiang Zhou
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Zhao Ren
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Michelle Chen
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Yeon-Hee Lim
- Merck & Co., Inc, 213 E. Grand Ave, South San Francisco, CA, 94080, USA
| | - Nathan G Hatcher
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Xiujuan Wen
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Payal R Sheth
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - David G McLaren
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Stephen F Previs
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| |
Collapse
|
4
|
Vachal P, Duffy JL, Campeau LC, Amin RP, Mitra K, Murphy BA, Shao PP, Sinclair PJ, Ye F, Katipally R, Lu Z, Ondeyka D, Chen YH, Zhao K, Sun W, Tyagarajan S, Bao J, Wang SP, Cote J, Lipardi C, Metzger D, Leung D, Hartmann G, Wollenberg GK, Liu J, Tan L, Xu Y, Chen Q, Liu G, Blaustein RO, Johns DG. Invention of MK-8262, a Cholesteryl Ester Transfer Protein (CETP) Inhibitor Backup to Anacetrapib with Best-in-Class Properties. J Med Chem 2021; 64:13215-13258. [PMID: 34375108 DOI: 10.1021/acs.jmedchem.1c00959] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cholesteryl ester transfer protein (CETP) represents one of the key regulators of the homeostasis of lipid particles, including high-density lipoprotein (HDL) and low-density lipoprotein (LDL) particles. Epidemiological evidence correlates increased HDL and decreased LDL to coronary heart disease (CHD) risk reduction. This relationship is consistent with a clinical outcomes trial of a CETP inhibitor (anacetrapib) combined with standard of care (statin), which led to a 9% additional risk reduction compared to standard of care alone. We discuss here the discovery of MK-8262, a CETP inhibitor with the potential for being the best-in-class molecule. Novel in vitro and in vivo paradigms were integrated to drug discovery to guide optimization informed by a critical understanding of key clinical adverse effect profiles. We present preclinical and clinical evidence of MK-8262 safety and efficacy by means of HDL increase and LDL reduction as biomarkers for reduced CHD risk.
Collapse
Affiliation(s)
- Petr Vachal
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Joseph L Duffy
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Louis-Charles Campeau
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Rupesh P Amin
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Kaushik Mitra
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Beth Ann Murphy
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Pengcheng P Shao
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Peter J Sinclair
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Feng Ye
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Revathi Katipally
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Zhijian Lu
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Debra Ondeyka
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Yi-Heng Chen
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Kake Zhao
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Wanying Sun
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Sriram Tyagarajan
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Jianming Bao
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Sheng-Ping Wang
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Josee Cote
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Concetta Lipardi
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Daniel Metzger
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Dennis Leung
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Georgy Hartmann
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Gordon K Wollenberg
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Jian Liu
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Lushi Tan
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Yingju Xu
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Qinghao Chen
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Guiquan Liu
- WuXi AppTec, 90 Delin Rd., Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Robert O Blaustein
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| | - Douglas G Johns
- Merck & Co., Inc., 2000 Galloping Hill Rd., Kenilworth, New Jersey 07033, United States
| |
Collapse
|
5
|
Jacome-Sosa M, Hu Q, Manrique-Acevedo CM, Phair RD, Parks EJ. Human intestinal lipid storage through sequential meals reveals faster dinner appearance is associated with hyperlipidemia. JCI Insight 2021; 6:e148378. [PMID: 34369385 PMCID: PMC8489663 DOI: 10.1172/jci.insight.148378] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Background It is increasingly recognized that intestinal cells can store lipids after a meal, yet the effect of this phenomenon on lipid absorption patterns in insulin resistance remains unknown. Methods The kinetics of meal fat appearance were measured in insulin-sensitive (IS, n = 8) and insulin-resistant (IR, n = 8) subjects after sequential, isotopically labeled lunch and dinner meals. Plasma dynamics on triacylglycerol-rich (TAG-rich) lipoproteins and plasma hormones were analyzed using a nonlinear, non–steady state kinetic model. Results At the onset of dinner, IS subjects showed an abrupt plasma appearance of lunch lipid consistent with the “second-meal effect,” followed by slower appearance of dinner fat in plasma, resulting in reduced accumulation of dinner TAG of 48% compared with lunch. By contrast, IR subjects exhibited faster meal TAG appearance rates after both lunch and dinner. This effect of lower enterocyte storage between meals was associated with greater nocturnal and next-morning hyperlipidemia. The biochemical data and the kinetic analysis of second-meal effect dynamics are consistent with rapid secretion of stored TAG bypassing lipolysis and resynthesis. In addition, the data are consistent with a role for the diurnal pattern of plasma leptin in regulating the processing of dietary lipid. Conclusion These data support the concept that intestinal lipid storage may be physiologically beneficial in IS subjects. Trial registration ClinicalTrials.gov NCT02020343. Funding This study was supported by a grant from the American Diabetes Association (grant 1-13-TS-12).
Collapse
Affiliation(s)
| | - Qiong Hu
- Department of Nutrition and Exercise Physiology and
| | | | - Robert D Phair
- Integrative Bioinformatics, Inc., Mountain View, California, USA
| | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology and.,Division of Gastroenterology and Hepatology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
6
|
Daurio NA, Wang Y, Chen Y, Zhou H, Carballo-Jane E, Mane J, Rodriguez CG, Zafian P, Houghton A, Addona G, McLaren DG, Zhang R, Shyong BJ, Bateman K, Downes DP, Webb M, Kelley DE, Previs SF. Spatial and temporal studies of metabolic activity: contrasting biochemical kinetics in tissues and pathways during fasted and fed states. Am J Physiol Endocrinol Metab 2019; 316:E1105-E1117. [PMID: 30912961 DOI: 10.1152/ajpendo.00459.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The regulation of nutrient homeostasis, i.e., the ability to transition between fasted and fed states, is fundamental in maintaining health. Since food is typically consumed over limited (anabolic) periods, dietary components must be processed and stored to counterbalance the catabolic stress that occurs between meals. Herein, we contrast tissue- and pathway-specific metabolic activity in fasted and fed states. We demonstrate that knowledge of biochemical kinetics that is obtained from opposite ends of the energetic spectrum can allow mechanism-based differentiation of healthy and disease phenotypes. Rat models of type 1 and type 2 diabetes serve as case studies for probing spatial and temporal patterns of metabolic activity via [2H]water labeling. Experimental designs that capture integrative whole body metabolism, including meal-induced substrate partitioning, can support an array of research surrounding metabolic disease; the relative simplicity of the approach that is discussed here should enable routine applications in preclinical models.
Collapse
Affiliation(s)
- Natalie A Daurio
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Yichen Wang
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Ying Chen
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Haihong Zhou
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Ester Carballo-Jane
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Joel Mane
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Carlos G Rodriguez
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Peter Zafian
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Andrea Houghton
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - George Addona
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - David G McLaren
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Rena Zhang
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Bao Jen Shyong
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Kevin Bateman
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Daniel P Downes
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Maria Webb
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - David E Kelley
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Stephen F Previs
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| |
Collapse
|
7
|
Lippincott-Schwartz J, Snapp EL, Phair RD. The Development and Enhancement of FRAP as a Key Tool for Investigating Protein Dynamics. Biophys J 2018; 115:1146-1155. [PMID: 30219286 DOI: 10.1016/j.bpj.2018.08.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/27/2018] [Accepted: 08/06/2018] [Indexed: 01/18/2023] Open
Abstract
The saga of fluorescence recovery after photobleaching (FRAP) illustrates how disparate technical developments impact science. Starting with the classic 1976 Axelrod et al. work in Biophysical Journal, FRAP (originally fluorescence photobleaching recovery) opened the door to extraction of quantitative information from photobleaching experiments, laying the experimental and theoretical groundwork for quantifying both the mobility and the mobile fraction of a labeled population of proteins. Over the ensuing years, FRAP's reach dramatically expanded, with new developments in GFP technology and turn-key confocal microscopy, which enabled measurement of protein diffusion and binding/dissociation rates in virtually every compartment within the cell. The FRAP technique and data catalyzed an exchange of ideas between biophysicists studying membrane dynamics, cell biologists focused on intracellular dynamics, and systems biologists modeling the dynamics of cell activity. The outcome transformed the field of cellular biology, leading to a fundamental rethinking of long-held theories of cellular dynamism. Here, we review the pivotal FRAP studies that made these developments and conceptual changes possible, which gave rise to current models of complex cell dynamics.
Collapse
Affiliation(s)
| | - Erik Lee Snapp
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia.
| | - Robert D Phair
- Integrative Bioinformatics, Inc., Mountain View, California
| |
Collapse
|
8
|
Previs SF, Herath K, Nawrocki AR, Rodriguez CG, Slipetz D, Singh SB, Kang L, Bhat G, Roddy TP, Conarello S, Terebetski J, Erion MD, Kelley DE. Using [ 2H]water to quantify the contribution of de novo palmitate synthesis in plasma: enabling back-to-back studies. Am J Physiol Endocrinol Metab 2018; 315:E63-E71. [PMID: 29351479 DOI: 10.1152/ajpendo.00010.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An increased contribution of de novo lipogenesis (DNL) may play a role in cases of dyslipidemia and adipose accretion; this suggests that inhibition of fatty acid synthesis may affect clinical phenotypes. Since it is not clear whether modulation of one step in the lipogenic pathway is more important than another, the use of tracer methods can provide a deeper level of insight regarding the control of metabolic activity. Although [2H]water is generally considered a reliable tracer for quantifying DNL in vivo (it yields a homogenous and quantifiable precursor labeling), the relatively long half-life of body water is thought to limit the ability of performing repeat studies in the same subjects; this can create a bottleneck in the development and evaluation of novel therapeutics for inhibiting DNL. Herein, we demonstrate the ability to perform back-to-back studies of DNL using [2H]water. However, this work uncovered special circumstances that affect the data interpretation, i.e., it is possible to obtain seemingly negative values for DNL. Using a rodent model, we have identified a physiological mechanism that explains the data. We show that one can use [2H]water to test inhibitors of DNL by performing back-to-back studies in higher species [i.e., treat nonhuman primates with platensimycin, an inhibitor of fatty acid synthase]; studies also demonstrate the unsuitability of [13C]acetate.
Collapse
Affiliation(s)
- Stephen F Previs
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Kithsiri Herath
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Andrea R Nawrocki
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Carlos G Rodriguez
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Deborah Slipetz
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Sheo B Singh
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Ling Kang
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Gowri Bhat
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Thomas P Roddy
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Stacey Conarello
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Jenna Terebetski
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Mark D Erion
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - David E Kelley
- Merck Research Laboratories, Merck & Company, Incorporated, Kenilworth, New Jersey
| |
Collapse
|
9
|
More VR, Lao J, McLaren DG, Cumiskey AM, Murphy BA, Chen Y, Previs S, Stout S, Patel R, Satapati S, Li W, Kowalik E, Szeto D, Nawrocki A, Pocai A, Wang L, Carrington P. Glucagon like receptor 1/ glucagon dual agonist acutely enhanced hepatic lipid clearance and suppressed de novo lipogenesis in mice. PLoS One 2017; 12:e0186586. [PMID: 29065174 PMCID: PMC5655430 DOI: 10.1371/journal.pone.0186586] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/03/2017] [Indexed: 02/08/2023] Open
Abstract
Lipid lowering properties of glucagon have been reported. Blocking glucagon signaling leads to rise in plasma LDL levels. Here, we demonstrate the lipid lowering effects of acute dosing with Glp1r/Gcgr dual agonist (DualAG). All the experiments were performed in 25 week-old male diet-induced (60% kCal fat) obese mice. After 2 hrs of fasting, mice were injected subcutaneously with vehicle, liraglutide (25nmol/kg) and DualAG (25nmol/kg). De novo cholesterol and palmitate synthesis was measured by deuterium incorporation method using D2O. 13C18-oleate infusion was used for measuring fatty acid esterification. Simultaneous activation of Glp1r and Gcgr resulted in decrease in plasma triglyceride and cholesterol levels. DualAG enhanced hepatic LDLr protein levels, along with causing decrease in content of plasma ApoB48 and ApoB100. VLDL secretion, de novo palmitate synthesis and fatty acid esterification decreased with acute DualAG treatment. On the other hand, ketone levels were elevated with DualAG treatment, indicating increased fatty acid oxidation. Lipid relevant changes were absent in liraglutide treated group. In an acute treatment, DualAG demonstrated significant impact on lipid homeostasis, specifically on hepatic uptake, VLDL secretion and de novo synthesis. These effects collectively reveal that lipid lowering abilities of DualAG are primarily through glucagon signaling and are liver centric.
Collapse
Affiliation(s)
- Vijay R. More
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Julie Lao
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - David G. McLaren
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Anne-Marie Cumiskey
- In-Vivo Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Beth Ann Murphy
- In-Vivo Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Ying Chen
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Stephen Previs
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Steven Stout
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Rajesh Patel
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Santhosh Satapati
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Wenyu Li
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Edward Kowalik
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Daphne Szeto
- In-Vivo Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Andrea Nawrocki
- In-Vivo Pharmacology, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Alessandro Pocai
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Liangsu Wang
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Paul Carrington
- Cardiometabolic Diseases Biology- Discovery, Merck & Co., Inc., Kenilworth, NJ, United States of America
| |
Collapse
|
10
|
Daurio NA, Wang SP, Chen Y, Zhou H, McLaren DG, Roddy TP, Johns DG, Milot D, Kasumov T, Erion MD, Kelley DE, Previs SF. Enhancing Studies of Pharmacodynamic Mechanisms via Measurements of Metabolic Flux: Fundamental Concepts and Guiding Principles for Using Stable Isotope Tracers. J Pharmacol Exp Ther 2017; 363:80-91. [PMID: 28724692 DOI: 10.1124/jpet.117.241091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 06/14/2017] [Indexed: 03/08/2025] Open
Abstract
Drug discovery and development efforts are largely based around a common expectation, namely, that direct or indirect action on a cellular process (e.g., statin-mediated enzyme inhibition or insulin-stimulated receptor activation) will have a beneficial impact on physiologic homeostasis. To expand on this, one could argue that virtually all pharmacologic interventions attempt to influence the flow of "traffic" in a biochemical network, irrespective of disease or modality. Since stable isotope tracer kinetic methods provide a measure of traffic flow (i.e., metabolic flux), their inclusion in study designs can yield novel information regarding pathway biology; the application of such methods requires the integration of knowledge in physiology, analytical chemistry, and mathematical modeling. Herein, we review the fundamental concepts that surround the use of tracer kinetics, define basic terms, and outline guiding principles via theoretical and experimental problems. Specifically, one needs to 1) recognize the types of biochemical events that change isotopic enrichments, 2) appreciate the distinction between fractional turnover and flux rate, and 3) be aware of the subtle differences between tracer kinetics and pharmacokinetics. We hope investigators can use the framework presented here to develop applications that address their specific questions surrounding biochemical flux, and thereby gain insight into the pathophysiology of disease states, and examine pharmacodynamic mechanisms.
Collapse
Affiliation(s)
- Natalie A Daurio
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Sheng-Ping Wang
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Ying Chen
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Haihong Zhou
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - David G McLaren
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Thomas P Roddy
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Douglas G Johns
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Denise Milot
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Takhar Kasumov
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Mark D Erion
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - David E Kelley
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| | - Stephen F Previs
- Merck & Company, Inc., Kenilworth, New Jersey (N.A.D., S.P.W., Y.C., H.Z., D.G.M., T.P.R., D.G.J., D.M.E., D.E.K., S.P.F.), and Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio (T.K.)
| |
Collapse
|
11
|
Millar JS, Lassman ME, Thomas T, Ramakrishnan R, Jumes P, Dunbar RL, deGoma EM, Baer AL, Karmally W, Donovan DS, Rafeek H, Wagner JA, Holleran S, Obunike J, Liu Y, Aoujil S, Standiford T, Gutstein DE, Ginsberg HN, Rader DJ, Reyes-Soffer G. Effects of CETP inhibition with anacetrapib on metabolism of VLDL-TG and plasma apolipoproteins C-II, C-III, and E. J Lipid Res 2017; 58:1214-1220. [PMID: 28314859 PMCID: PMC5454510 DOI: 10.1194/jlr.m074880] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/16/2017] [Indexed: 01/30/2023] Open
Abstract
Cholesteryl ester transfer protein (CETP) mediates the transfer of HDL cholesteryl esters for triglyceride (TG) in VLDL/LDL. CETP inhibition, with anacetrapib, increases HDL-cholesterol, reduces LDL-cholesterol, and lowers TG levels. This study describes the mechanisms responsible for TG lowering by examining the kinetics of VLDL-TG, apoC-II, apoC-III, and apoE. Mildly hypercholesterolemic subjects were randomized to either placebo (N = 10) or atorvastatin 20 mg/qd (N = 29) for 4 weeks (period 1) followed by 8 weeks of anacetrapib, 100 mg/qd (period 2). Following each period, subjects underwent stable isotope metabolic studies to determine the fractional catabolic rates (FCRs) and production rates (PRs) of VLDL-TG and plasma apoC-II, apoC-III, and apoE. Anacetrapib reduced the VLDL-TG pool on a statin background due to an increased VLDL-TG FCR (29%; P = 0.002). Despite an increased VLDL-TG FCR following anacetrapib monotherapy (41%; P = 0.11), the VLDL-TG pool was unchanged due to an increase in the VLDL-TG PR (39%; P = 0.014). apoC-II, apoC-III, and apoE pool sizes increased following anacetrapib; however, the mechanisms responsible for these changes differed by treatment group. Anacetrapib increased the VLDL-TG FCR by enhancing the lipolytic potential of VLDL, which lowered the VLDL-TG pool on atorvastatin background. There was no change in the VLDL-TG pool in subjects treated with anacetrapib monotherapy due to an accompanying increase in the VLDL-TG PR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Joseph Obunike
- New York City College of Technology, CUNY, Brooklyn, NY 11201
| | - Yang Liu
- Merck & Co., Inc., Kenilworth, NJ 07033
| | | | | | | | | | | | | |
Collapse
|