1
|
Lee H, Im W. Substrates (Acyl-CoA and Diacylglycerol) Entry and Products (CoA and Triacylglycerol) Egress Pathways in DGAT1. J Comput Chem 2025; 46:e70108. [PMID: 40251888 PMCID: PMC12008735 DOI: 10.1002/jcc.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/13/2025] [Accepted: 04/05/2025] [Indexed: 04/21/2025]
Abstract
Diacylglycerol O-acyltransferase 1 (DGAT1) is an integral membrane protein that uses acyl-coenzyme A (acyl-CoA) and diacylglycerol (DAG) to catalyze the formation of triacylglycerides (TAGs). The acyl transfer reaction occurs between the activated carboxylate group of the fatty acid and the free hydroxyl group on the glycerol backbone of DAG. However, how the two substrates enter DGAT1's catalytic reaction chamber and interact with DGAT1 remains elusive. This study aims to explore the structural basis of DGAT1's substrate recognition by investigating each substrate's pathway to the reaction chamber. Using a human DGAT1 cryo-EM structure in complex with an oleoyl-CoA substrate, we designed two different all-atom molecular dynamics (MD) simulation systems: DGAT1away (both acyl-CoA and DAG away from the reaction chamber) and DGAT1bound (acyl-CoA bound in and DAG away from the reaction chamber). Our DGAT1away simulations reveal that acyl-CoA approaches the reaction chamber via interactions with positively charged residues in transmembrane helix 7. DGAT1bound simulations show DAGs entering into the reaction chamber from the cytosol leaflet. The bound acyl-CoA's fatty acid lines up with the headgroup of DAG, which appears to be competent to TAG formation. We then converted them into TAG and coenzyme (CoA) and used adaptive biasing force (ABF) simulations to explore the egress pathways of the products. We identify their escape routes, which are aligned with their respective entry pathways. Visualization of the substrate and product pathways and their interactions with DGAT1 is expected to guide future experimental design to better understand DGAT1 structure and function.
Collapse
Affiliation(s)
- Hwayoung Lee
- Department of Biological SciencesLehigh UniversityBethlehemPennsylvaniaUSA
| | - Wonpil Im
- Department of Biological SciencesLehigh UniversityBethlehemPennsylvaniaUSA
| |
Collapse
|
2
|
Li P, Jiang W. A New Insight on Atherosclerosis Mechanism and Lipid-Lowering Drugs. Rev Cardiovasc Med 2025; 26:25321. [PMID: 40160588 PMCID: PMC11951287 DOI: 10.31083/rcm25321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 04/02/2025] Open
Abstract
Atherosclerosis (AS) is a chronic vascular disease primarily affecting large and medium-sized arteries, involving complex pathological mechanisms such as inflammatory responses, lipid metabolism disorders and vascular plaque formation. In recent years, several emerging research hotspots have appeared in the field of atherosclerosis, including gut microbiota, pyroptosis, ferroptosis, autophagy, cuproptosis, exosomes and non-coding RNA. Traditional lipid-lowering drugs play a crucial role in the treatment of AS but are not able to significantly reverse the pathological changes. This article aims to summarize the latest research progress in the pathogenesis of AS and the diagnosis and treatment of the disease by comprehensively analyzing relevant literature mainly from the past five years. Additionally, the mechanisms of action and research advances of statins, cholesterol absorption inhibitors, fibrates and novel lipid-lowering drugs are reviewed to provide new insights into the diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Penghui Li
- Binhai New Area Hospital of TCM, 300000 Tianjin, China
| | - Wei Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300000 Tianjin, China
| |
Collapse
|
3
|
Oleszycka E, Kwiecień K, Grygier B, Cichy J, Kwiecińska P. The many faces of DGAT1. Life Sci 2025; 362:123322. [PMID: 39709166 DOI: 10.1016/j.lfs.2024.123322] [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: 09/24/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) is a multifaced enzyme with a wide spectrum of substrates, from lipids through waxes to retinoids, which makes it an interesting therapeutic target. DGAT1 inhibitors are currently at various stages of preclinical and clinical trials, mostly related to metabolic diseases. Interestingly, in recent years, a growing amount of research has shown the influence of DGAT1 on immune cell metabolism and functions, highlighting its important role during infections and tumorigenesis. In this review, we aim to elucidate the potential immunomodulatory effect of DGAT1 in physiological and pathological conditions.
Collapse
Affiliation(s)
- Ewa Oleszycka
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Kamila Kwiecień
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Beata Grygier
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Science, Cracow, Poland
| | - Joanna Cichy
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Patrycja Kwiecińska
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland; Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland.
| |
Collapse
|
4
|
Wang L, Xu S, Zhou M, Hu H, Li J. The role of DGAT1 and DGAT2 in tumor progression via fatty acid metabolism: A comprehensive review. Int J Biol Macromol 2024; 278:134835. [PMID: 39154689 DOI: 10.1016/j.ijbiomac.2024.134835] [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: 04/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Fatty acid metabolism is a complex biochemical process, including the production, breakdown and application of fatty acids. Not only is it an important component of lipid metabolism, fatty acid metabolism is also connected to the energy metabolism pathways of cells and plays a vital role in maintaining the energy balance of organisms. Diacylglycerol-O-acyltransferase 1 (DGAT1) and Diacylglycerol-O-acyltransferase 2 (DGAT2) are key components in regulating lipid metabolism, which provide energy for cell proliferation and growth. Recent studies have shown that DGAT1 and DGAT2 influence tumor progression through fatty acid metabolism in cancer. Although DGAT1 and DGAT2 have similar names, they differ significantly in various aspects and play distinct roles in individual tumors. A comparative analysis of the physiological roles of these enzymes and their differential expressions in different types of tumors will enhance our understanding of their unique characteristics. This article summarizes the characteristics of tumor fatty acid metabolism and explains how DGAT1 and DGAT2 specifically promote tumor progression. In addition, this review discusses the potential of lipid-lowering drugs in tumor treatment, providing a new perspective on targeting fatty acid metabolism to inhibit tumor progression in the future, while emphasizing the importance of DGAT1 and DGAT2 as potential targets for tumor treatment.
Collapse
Affiliation(s)
- Leisheng Wang
- Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Shiwei Xu
- Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Mengzhen Zhou
- Southeast University School of Medicine, Nanjing 210009, China
| | - Hao Hu
- Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China.
| | - Jinyou Li
- Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
5
|
Longo M, Paolini E, Di Benedetto P, Tomassini E, Meroni M, Dongiovanni P. DGAT1 and DGAT2 Inhibitors for Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Management: Benefits for Their Single or Combined Application. Int J Mol Sci 2024; 25:9074. [PMID: 39201759 PMCID: PMC11354429 DOI: 10.3390/ijms25169074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Inhibiting diacylglycerol acetyltransferase (DGAT1, DGAT2) enzymes (iDGAT1, iDGAT2), involved in triglyceride (TG) synthesis, improves hepatic steatosis in metabolic dysfunction-associated steatotic liver disease (MASLD) patients. However, their potential synergism in disease onset (SLD) and progression (metabolic dysfunction-associated steatohepatitis, fibrosis) has been poorly explored. We investigated iDGAT1 and iDGAT2 efficacy, alone or combined (iDGAT1/2) on fat accumulation and hepatocellular injury in hepatocytes (HepG2) and on fibrogenic processes in hepatic stellate cells (LX2). We further tested whether the addition of MitoQ antioxidant to iDGAT1/2 would enhance their effects. SLD and MASH conditions were reproduced in vitro by supplementing Dulbecco's Modified Eagle's Medium (DMEM) with palmitic/oleic acids (PAOA) alone (SLD-medium), or plus Lipopolisaccaride (LPS), fructose, and glucose (MASH-medium). In SLD-medium, iDGAT1 and iDGAT2 individually, and even more in combination, reduced TG synthesis in HepG2 cells. Markers of hepatocellular damage were slightly decreased after single iDGAT exposure. Conversely, iDGAT1/2 counteracted ER/oxidative stress and inflammation and enhanced mitochondrial Tricarboxylic acid cycle (TCA) and respiration. In HepG2 cells under a MASH-like condition, only iDGAT1/2 effectively ameliorated TG content and oxidative and inflammatory mediators, further improving bioenergetic balance. LX2 cells, challenged with SLD/MASH media, showed less proliferation and slower migration rates in response to iDGAT1/2 drugs. MitoQ combined with iDGAT1/2 improved cell viability and dampened free fatty acid release by stimulating β-oxidation. Dual DGAT inhibition combined with antioxidants open new perspectives for MASLD management.
Collapse
Affiliation(s)
| | | | | | | | | | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.L.); (E.P.); (P.D.B.); (E.T.); (M.M.)
| |
Collapse
|
6
|
Deng B, Kong W, Shen X, Han C, Zhao Z, Chen S, Zhou C, Bae-Jump V. The role of DGAT1 and DGAT2 in regulating tumor cell growth and their potential clinical implications. J Transl Med 2024; 22:290. [PMID: 38500157 PMCID: PMC10946154 DOI: 10.1186/s12967-024-05084-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/10/2024] [Indexed: 03/20/2024] Open
Abstract
Lipid metabolism is widely reprogrammed in tumor cells. Lipid droplet is a common organelle existing in most mammal cells, and its complex and dynamic functions in maintaining redox and metabolic balance, regulating endoplasmic reticulum stress, modulating chemoresistance, and providing essential biomolecules and ATP have been well established in tumor cells. The balance between lipid droplet formation and catabolism is critical to maintaining energy metabolism in tumor cells, while the process of energy metabolism affects various functions essential for tumor growth. The imbalance of synthesis and catabolism of fatty acids in tumor cells leads to the alteration of lipid droplet content in tumor cells. Diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2, the enzymes that catalyze the final step of triglyceride synthesis, participate in the formation of lipid droplets in tumor cells and in the regulation of cell proliferation, migration and invasion, chemoresistance, and prognosis in tumor. Several diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2 inhibitors have been developed over the past decade and have shown anti-tumor effects in preclinical tumor models and improvement of metabolism in clinical trials. In this review, we highlight key features of fatty acid metabolism and different paradigms of diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2 activities on cell proliferation, migration, chemoresistance, and prognosis in tumor, with the hope that these scientific findings will have potential clinical implications.
Collapse
Affiliation(s)
- Boer Deng
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiaochang Shen
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chao Han
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
| | - Ziyi Zhao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shuning Chen
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
7
|
Stallard L, Siddiqui I, Muise A. Beyond IBD: the genetics of other early-onset diarrhoeal disorders. Hum Genet 2023; 142:655-667. [PMID: 36788146 PMCID: PMC10182111 DOI: 10.1007/s00439-023-02524-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 01/19/2023] [Indexed: 02/16/2023]
Abstract
Diarrhoeal disorders in childhood extend beyond the inflammatory bowel diseases. Persistent and severe forms of diarrhoea can occur from birth and are associated with significant morbidity and mortality. These disorders can affect not only the gastrointestinal tract but frequently have extraintestinal manifestations, immunodeficiencies and endocrinopathies. Genomic analysis has advanced our understanding of these conditions and has revealed precision-based treatment options such as potentially curative haematopoietic stem cell transplant. Although many new mutations have been discovered, there is frequently no clear genotype-phenotype correlation. The functional effects of gene mutations can be studied in model systems such as patient-derived organoids. This allows us to further characterise these disorders and advance our understanding of the pathophysiology of the intestinal mucosa. In this review, we will provide an up to date overview of genes involved in diarrhoeal disorders of early onset, particularly focussing on the more recently described gene defects associated with protein loosing enteropathy.
Collapse
Affiliation(s)
- Lorraine Stallard
- SickKids Inflammatory Bowel Disease Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Iram Siddiqui
- Division of Pathology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Aleixo Muise
- SickKids Inflammatory Bowel Disease Centre, The Hospital for Sick Children, Toronto, ON, Canada. .,Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Pediatrics, Institute of Medical Science and Biochemistry, University of Toronto, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
8
|
Chen G, Harwood JL, Lemieux MJ, Stone SJ, Weselake RJ. Acyl-CoA:diacylglycerol acyltransferase: Properties, physiological roles, metabolic engineering and intentional control. Prog Lipid Res 2022; 88:101181. [PMID: 35820474 DOI: 10.1016/j.plipres.2022.101181] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022]
Abstract
Acyl-CoA:diacylglycerol acyltransferase (DGAT, EC 2.3.1.20) catalyzes the last reaction in the acyl-CoA-dependent biosynthesis of triacylglycerol (TAG). DGAT activity resides mainly in membrane-bound DGAT1 and DGAT2 in eukaryotes and bifunctional wax ester synthase-diacylglycerol acyltransferase (WSD) in bacteria, which are all membrane-bound proteins but exhibit no sequence homology to each other. Recent studies also identified other DGAT enzymes such as the soluble DGAT3 and diacylglycerol acetyltransferase (EaDAcT), as well as enzymes with DGAT activities including defective in cuticular ridges (DCR) and steryl and phytyl ester synthases (PESs). This review comprehensively discusses research advances on DGATs in prokaryotes and eukaryotes with a focus on their biochemical properties, physiological roles, and biotechnological and therapeutic applications. The review begins with a discussion of DGAT assay methods, followed by a systematic discussion of TAG biosynthesis and the properties and physiological role of DGATs. Thereafter, the review discusses the three-dimensional structure and insights into mechanism of action of human DGAT1, and the modeled DGAT1 from Brassica napus. The review then examines metabolic engineering strategies involving manipulation of DGAT, followed by a discussion of its therapeutic applications. DGAT in relation to improvement of livestock traits is also discussed along with DGATs in various other eukaryotic organisms.
Collapse
Affiliation(s)
- Guanqun Chen
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada.
| | - John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Membrane Protein Disease Research Group, Edmonton T6G 2H7, Canada
| | - Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| | - Randall J Weselake
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada
| |
Collapse
|
9
|
Ghanem M, Lewis GF, Xiao C. Recent advances in cytoplasmic lipid droplet metabolism in intestinal enterocyte. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159197. [PMID: 35820577 DOI: 10.1016/j.bbalip.2022.159197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 06/03/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
Processing of dietary fats in the intestine is a highly regulated process that influences whole-body energy homeostasis and multiple physiological functions. Dysregulated lipid handling in the intestine leads to dyslipidemia and atherosclerotic cardiovascular disease. In intestinal enterocytes, lipids are incorporated into lipoproteins and cytoplasmic lipid droplets (CLDs). Lipoprotein synthesis and CLD metabolism are inter-connected pathways with multiple points of regulation. This review aims to highlight recent advances in the regulatory mechanisms of lipid processing in the enterocyte, with particular focus on CLDs. In-depth understanding of the regulation of lipid metabolism in the enterocyte may help identify therapeutic targets for the treatment and prevention of metabolic disorders.
Collapse
Affiliation(s)
- Murooj Ghanem
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Gary F Lewis
- Departments of Medicine and Physiology, University of Toronto, and University Health Network, Toronto, ON, Canada
| | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
10
|
Abstract
INTRODUCTION Primary chylomicronemia is characterized by pathological accumulation of chylomicrons in the plasma causing severe hypertriglyceridemia, typically >10 mmol/L (>875 mg/dL). Patients with the ultra-rare familial chylomicronemia syndrome (FCS) subtype completely lack lipolytic capacity and respond minimally to traditional triglyceride-lowering therapies. The mainstay of treatment is a low-fat diet, which is difficult to follow and compromises quality of life. New therapies are being developed primarily to prevent episodes of life-threatening acute pancreatitis. AREAS COVERED Antagonists of apolipoprotein (apo) C-III, such as the antisense oligonucleotide (ASO) volanesorsen, significantly reduce triglyceride levels in chylomicronemia. However, approval of and access to volanesorsen are restricted since a substantial proportion of treated FCS patients developed thrombocytopenia. Newer apo C-III antagonists, namely, the ASO olezarsen (formerly AKCEA-APOCIII-LRx) and short interfering RNA (siRNA) ARO-APOC3, appear to show efficacy with less risk of thrombocytopenia. Potential utility of antagonists of angiopoietin-like protein 3 (ANGPTL3) such as evinacumab and the siRNA ARO-ANG3 in subtypes of chylomicronemia remains to be defined. EXPERT OPINION Emerging pharmacologic therapies for chylomicronemia show promise, particularly apo C-III antagonists. However, these treatments are still investigational. Further study of their efficacy and safety in patients with both rare FCS and more common multifactorial chylomicronemia is needed.
Collapse
Affiliation(s)
- Isabel Shamsudeen
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Robert A Hegele
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
11
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 475] [Impact Index Per Article: 158.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
12
|
Stone SJ. Mechanisms of intestinal triacylglycerol synthesis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159151. [PMID: 35296424 DOI: 10.1016/j.bbalip.2022.159151] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/13/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023]
Abstract
Triacylglycerols are a major source of stored energy that are obtained either from the diet or can be synthesized to some extent by most tissues. Alterations in pathways of triacylglycerol metabolism can result in their excessive accumulation leading to obesity, insulin resistance, cardiovascular disease and nonalcoholic fatty liver disease. Most tissues in mammals synthesize triacylglycerols via the glycerol 3-phosphate pathway. However, in the small intestine the monoacylglycerol acyltransferase pathway is the predominant pathway for triacylglycerol biosynthesis where it participates in the absorption of dietary triacylglycerol. In this review, the enzymes that are part of both the glycerol 3-phosphate and monoacylglycerol acyltransferase pathways and their contributions to intestinal triacylglycerol metabolism are reviewed. The potential of some of the enzymes involved in triacylglycerol synthesis in the small intestine as possible therapeutic targets for treating metabolic disorders associated with elevated triacylglycerol is briefly discussed.
Collapse
Affiliation(s)
- Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
13
|
Managing of Dyslipidaemia Characterized by Accumulation of Triglyceride-Rich Lipoproteins. Curr Atheroscler Rep 2022; 24:1-12. [PMID: 35107764 PMCID: PMC8924084 DOI: 10.1007/s11883-022-00979-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2021] [Indexed: 11/05/2022]
Abstract
Purpose of Review The accumulation of triglyceride-rich lipoproteins (TRLs) in plasma in patients with familial chylomicronaemia syndrome (FCS) or severe hypertriglyceridemia is associated with an increased risk of potentially life-threatening pancreatitis. Elevated TRL levels have also been suggested to contribute to atherosclerotic cardiovascular disease (ASCVD). This review provides the latest progress that has been made in this field of research. Recent Findings Apolipoprotein C-III and angiopoietin-like protein 3 play key roles in the metabolism of TRLs. Targeting their production in the liver or their presence in the circulation effectively reduces triglycerides in patients with FCS or severe hypertriglyceridemia. Attempts to reduce triglyceride synthesis in the small intestine have been halted. Early studies with a fibroblast growth factor 21 agonist have shown to reduce plasma triglycerides and hepatic steatosis and improve glucose homeostasis. Summary New drugs have recently been shown to effectively reduce plasma triglycerides which render hope for treating the risk of pancreatitis. Studies that have just been initiated will learn whether this unmet clinical will be met. It is too early to evaluate the potential of these drugs to reduce the risk of atherosclerosis through the reduction of triglycerides.
Collapse
|
14
|
Laufs U, Parhofer KG, Ginsberg HN, Hegele RA. Clinical review on triglycerides. Eur Heart J 2021; 41:99-109c. [PMID: 31764986 PMCID: PMC6938588 DOI: 10.1093/eurheartj/ehz785] [Citation(s) in RCA: 318] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/23/2022] Open
Abstract
Hypertriglyceridaemia is a common clinical problem. Epidemiologic and genetic studies have established that triglyceride-rich lipoproteins (TRL) and their remnants as important contributors to ASCVD while severe hypertriglyceridaemia raises risk of pancreatitis. While low-density lipoprotein is the primary treatment target for lipid lowering therapy, secondary targets that reflect the contribution of TRL such as apoB and non-HDL-C are recommended in the current guidelines. Reduction of severely elevated triglycerides is important to avert or reduce the risk of pancreatitis. Here we discuss interventions for hypertriglyceridaemia, including diet and lifestyle, established treatments such as fibrates and omega-3 fatty acid preparations and emerging therapies, including various biological agents. ![]()
Collapse
Affiliation(s)
- Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Liebigstr. 20, Leipzig, Germany
| | - Klaus G Parhofer
- University Munich, Medical Department 4 - Grosshadern, Marchioninistr. 15, Munich, Germany
| | - Henry N Ginsberg
- Irving Institute for Clinical and Translational Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, USA
| | - Robert A Hegele
- Department of Medicine, Robarts Research Institute, Western University, London, Ontario, Canada
| |
Collapse
|
15
|
Okazaki H, Gotoda T, Ogura M, Ishibashi S, Inagaki K, Daida H, Hayashi T, Hori M, Masuda D, Matsuki K, Yokoyama S, Harada-Shiba M. Current Diagnosis and Management of Primary Chylomicronemia. J Atheroscler Thromb 2021; 28:883-904. [PMID: 33980761 PMCID: PMC8532063 DOI: 10.5551/jat.rv17054] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Primary chylomicronemia (PCM) is a rare and intractable disease characterized by marked accumulation of chylomicrons in plasma. The levels of plasma triglycerides (TGs) typically range from 1,000 - 15,000 mg/dL or higher.
PCM is caused by defects in the lipoprotein lipase (LPL) pathway due to genetic mutations, autoantibodies, or unidentified causes. The monogenic type is typically inherited as an autosomal recessive trait with loss-of-function mutations in LPL pathway genes (
LPL
,
LMF1
,
GPIHBP1
,
APOC2
, and
APOA5
). Secondary/environmental factors (diabetes, alcohol intake, pregnancy, etc.) often exacerbate hypertriglyceridemia (HTG).
The signs, symptoms, and complications of chylomicronemia include eruptive xanthomas, lipemia retinalis, hepatosplenomegaly, and acute pancreatitis with onset as early as in infancy. Acute pancreatitis can be fatal and recurrent episodes of abdominal pain may lead to dietary fat intolerance and failure to thrive. The main goal of treatment is to prevent acute pancreatitis by reducing plasma TG levels to at least less than 500-1,000 mg/dL. However, current TG-lowering medications are generally ineffective for PCM. The only other treatment options are modulation of secondary/environmental factors. Most patients need strict dietary fat restriction, which is often difficult to maintain and likely affects their quality of life. Timely diagnosis is critical for the best prognosis with currently available management, but PCM is often misdiagnosed and undertreated. The aim of this review is firstly to summarize the pathogenesis, signs, symptoms, diagnosis, and management of PCM, and secondly to propose simple diagnostic criteria that can be readily translated into general clinical practice to improve the diagnostic rate of PCM. In fact, these criteria are currently used to define eligibility to receive social support from the Japanese government for PCM as a rare and intractable disease. Nevertheless, further research to unravel the molecular pathogenesis and develop effective therapeutic modalities is warranted. Nationwide registry research on PCM is currently ongoing in Japan with the aim of better understanding the disease burden as well as the unmet needs of this life-threatening disease with poor therapeutic options.
Collapse
Affiliation(s)
- Hiroaki Okazaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Takanari Gotoda
- Department of Metabolic Biochemistry, Faculty of Medicine, Kyorin University
| | - Masatsune Ogura
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University
| | - Kyoko Inagaki
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Nippon Medical School
| | - Hiroyuki Daida
- Faculty of Health Science, Juntendo University, Juntendo University Graduate School of Medicine
| | - Toshio Hayashi
- School of Health Sciences, Nagoya University Graduate School of Medicine
| | - Mika Hori
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University
| | - Daisaku Masuda
- Department of Cardiology, Health Care Center, Rinku Innovation Center for Wellness Care and Activities (RICWA), Rinku General Medical Center
| | - Kota Matsuki
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine
| | | | - Mariko Harada-Shiba
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center Research Institute
| | | |
Collapse
|
16
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
17
|
Tomlinson B, Chan P, Zhang Y, Liu Z, Lam CWK. Pharmacokinetics of current and emerging treatments for hypercholesterolemia. Expert Opin Drug Metab Toxicol 2020; 16:371-385. [PMID: 32223657 DOI: 10.1080/17425255.2020.1749261] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Reduction of low-density-lipoprotein cholesterol (LDL-C) and other apolipoprotein B (apoB)-containing lipoproteins reduces cardiovascular (CV) events and greater reductions have greater benefits. Current lipid treatments cannot always achieve desirable LDL-C targets and additional or alternative treatments are often needed.Areas covered: In this article, we review the pharmacokinetics of the available and emerging treatments for hypercholesterolemia and focus on recently approved drugs and those at a late stage of development.Expert opinion: Statin pharmacokinetics are well known and appropriate drugs and doses can usually be chosen for individual patients to achieve LDL-C targets and avoid adverse effects and drug-drug interactions. Ezetimibe, icosapent ethyl and the monoclonal antibodies evolocumab and alirocumab have established efficacy and safety. Newer oral agents including pemafibrate and bempedoic acid have generally favorable pharmacokinetics supporting use in a wide range of patients. RNA-based therapies with antisense oligonucleotides are highly specific for their targets and those inhibiting apoB, apoCIII, angiopoietin-like protein 3 and lipoprotein(a) have shown promising results. The small-interfering RNA inclisiran has the notable advantage that a single subcutaneous administration may be effective for up to 6 months. The CV outcome trial results and long term safety data are eagerly awaited for these new agents.
Collapse
Affiliation(s)
- Brian Tomlinson
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan.,Research Center for Translational Medicine, Shanghai East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yuzhen Zhang
- Research Center for Translational Medicine, Shanghai East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, Shanghai East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | | |
Collapse
|
18
|
Florentin M, Kostapanos MS, Anagnostis P, Liamis G. Recent developments in pharmacotherapy for hypertriglyceridemia: what's the current state of the art? Expert Opin Pharmacother 2020; 21:107-120. [PMID: 31738617 DOI: 10.1080/14656566.2019.1691523] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/07/2019] [Indexed: 10/25/2022]
Abstract
Introduction: Hypertriglyceridemia is associated with both the development of cardiovascular disease (CVD) when mild-to-moderate and high risk of pancreatitis when more severe. The residual CVD risk after low-density lipoprotein cholesterol (LDL-C) lowering is, in part, attributed to high triglyceride (TG) levels. Therefore, there appears to be a need for effective TG-lowering agents.Areas covered: This review presents the most recent advances in hypertriglyceridemia treatment; specifically, it discusses the results of clinical trials and critically comments on apolipoprotein C-III inhibitors, angiopoietin-like 3 inhibitors, alipogene tiparvovec, pradigastat, pemafibrate and novel formulations of omega-3 fatty acids.Expert opinion: In the era of extreme lowering of LDL-C levels with several agents, there seems to be space for novel therapeutic options to combat parameters responsible for residual CVD risk, among which are elevated TGs. Furthermore, a significant number of individuals have very high TG levels and encounter the risk of acute pancreatitis. The most recently developed TG-lowering drugs appear to have a role in both conditions; the choice is mainly based on baseline TG levels. Dyslipidemia guidelines are likely to change in the near future to include some of these agents. Of course, long-term data regarding their safety and efficacy in terms of CVD outcomes and pancreatitis are warranted.
Collapse
Affiliation(s)
- Matilda Florentin
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Michael S Kostapanos
- Lipid clinic, Department of General Medicine, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Panagiotis Anagnostis
- Unit of reproductive endocrinology, 1st Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George Liamis
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
19
|
Abstract
Several new or emerging drugs for dyslipidemia owe their existence, in part, to human genetic evidence, such as observations in families with rare genetic disorders or in Mendelian randomization studies. Much effort has been directed to agents that reduce LDL (low-density lipoprotein) cholesterol, triglyceride, and Lp[a] (lipoprotein[a]), with some sustained programs on agents to raise HDL (high-density lipoprotein) cholesterol. Lomitapide, mipomersen, AAV8.TBG.hLDLR, inclisiran, bempedoic acid, and gemcabene primarily target LDL cholesterol. Alipogene tiparvovec, pradigastat, and volanesorsen primarily target elevated triglycerides, whereas evinacumab and IONIS-ANGPTL3-LRx target both LDL cholesterol and triglyceride. IONIS-APO(a)-LRx targets Lp(a).
Collapse
Affiliation(s)
- Robert A Hegele
- From the Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, Vascular Medicine Program, University of California San Diego, La Jolla (S.T.)
| |
Collapse
|
20
|
Abstract
An increased risk of cardiovascular disease, independent of conventional risk factors, is present even at minor levels of renal impairment and is highest in patients with end-stage renal disease (ESRD) requiring dialysis. Renal dysfunction changes the level, composition and quality of blood lipids in favour of a more atherogenic profile. Patients with advanced chronic kidney disease (CKD) or ESRD have a characteristic lipid pattern of hypertriglyceridaemia and low HDL cholesterol levels but normal LDL cholesterol levels. In the general population, a clear relationship exists between LDL cholesterol and major atherosclerotic events. However, in patients with ESRD, LDL cholesterol shows a negative association with these outcomes at below average LDL cholesterol levels and a flat or weakly positive association with mortality at higher LDL cholesterol levels. Overall, the available data suggest that lowering of LDL cholesterol is beneficial for prevention of major atherosclerotic events in patients with CKD and in kidney transplant recipients but is not beneficial in patients requiring dialysis. The 2013 Kidney Disease: Improving Global Outcomes (KDIGO) Clinical Practice Guideline for Lipid Management in CKD provides simple recommendations for the management of dyslipidaemia in patients with CKD and ESRD. However, emerging data and novel lipid-lowering therapies warrant some reappraisal of these recommendations.
Collapse
|
21
|
Harrison TJ, Bauer D, Berdichevsky A, Chen X, Duvadie R, Hoogheem B, Hatsis P, Liu Q, Mao J, Miduturu V, Rocheford E, Zecri F, Zessis R, Zheng R, Zhu Q, Streeper R, Patel SJ. Successful Strategies for Mitigation of a Preclinical Signal for Phototoxicity in a DGAT1 Inhibitor. ACS Med Chem Lett 2019; 10:1128-1133. [PMID: 31413796 DOI: 10.1021/acsmedchemlett.9b00117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/20/2019] [Indexed: 12/25/2022] Open
Abstract
Diacylglycerol O-acyltransferase 1 (DGAT1) inhibitor Pradigastat (1) was shown to be effective at decreasing postprandial triglyceride levels in a patient population with familial chylomicronemia syndrome (FCS). Although pradigastat does not cause photosensitization in humans at the high clinical dose of 40 mg, a positive signal was observed in preclinical models of phototoxicity. Herein, we describe a preclinical phototoxicity mitigation strategy for diarylamine containing molecules utilizing the introduction of an amide or suitable heterocyclic function. This strategy led to the development of two second-generation compounds with low risk of phototoxicity, disparate exposure profiles, and comparable efficacy to 1 in a rodent lipid bolus model for post-prandial plasma triglycerides.
Collapse
Affiliation(s)
| | - Daniel Bauer
- Preclinical Safety, Novartis Institutes for Biomedical Research, 4002 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Okour M, Gress A, Zhu X, Rieman D, Lickliter JD, Brigandi RA. First-in-Human Pharmacokinetics and Safety Study of GSK3008356, a Selective DGAT1 Inhibitor, in Healthy Volunteers. Clin Pharmacol Drug Dev 2019; 8:1088-1099. [PMID: 30950565 DOI: 10.1002/cpdd.691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 03/19/2019] [Indexed: 11/10/2022]
Abstract
Diacylglycerol acyltransferase (DGAT) enzymes are involved in triglyceride (TG) biosynthesis. GSK3008356 is a potent and selective DGAT1 inhibitor that was administered orally in a 2-part study as double-blind, randomized, placebo-controlled single doses (SDs) and repeat doses (RDs) in healthy subjects to investigate its pharmacokinetics, pharmacodynamics, and safety/tolerability. Gastrointestinal adverse events were considered drug related and increased with dose and when given as multiple doses. In the SD part (n = 80), GSK3008356 was dosed from 5 to 200 mg as single or multiple doses per day. In the RD part (n = 24), GSK3008356 was dosed twice daily at 1, 3, and 10 mg for 14 days. GSK3008356 was generally well tolerated in the SD and RD parts. With single doses, absorption was rapid (median tmax , 0.5-1.5 hours), whereas single-day divided dosing resulted in higher tmax . Following 14-day RD oral administration, GSK3008356 was also rapidly absorbed, with median tmax ranging from 0.5 to 0.75 hours on days 1 and 14. Estimated mean half-life ranged from 1.5 to 4.6 hours with SDs and 1.3 to 2.1 hours with RDs. Exposure of GSK3008356 was largely dose proportional after RDs. At higher doses, there was a trend toward lower absolute postprandial TG level in some subjects.
Collapse
|
23
|
Okour M, Brigandi RA, Tenero D. A population analysis of the DGAT1 inhibitor GSK3008356 and its effect on endogenous and meal-induced triglyceride turnover in healthy subjects. Fundam Clin Pharmacol 2019; 33:567-580. [PMID: 30790345 DOI: 10.1111/fcp.12455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/11/2019] [Accepted: 02/18/2019] [Indexed: 01/11/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a liver disease in which fatty infiltration is accompanied by liver inflammation. GSK3008356 is under development as a selective inhibitor of diacylglycerol acyltransferase 1 (DGAT1), a key enzyme involved in the formation of triglyceride (TG). Decreased DGAT1 activity can reduce circulating TG and liver TG, and therefore could potentially prevent or treat NASH. The aim of the current study was to develop a population pharmacokinetic-pharmacodynamic (PKPD) model that characterizes the PK disposition of GSK3008356 and its relation to the changes in blood TG. Drug concentrations were measured in 104 healthy adults receiving various single (SD) and repeat doses (RD) in a first time in human (FiH) study. A 30% fat meal was given at hour 2 postdose, and blood postprandial TG concentrations were measured at various time points. The population PKPD model consists of several parts including a PK model, drug effect model, meal effect model, and a turnover model. The pharmacokinetic data were described using a 3-compartment model. Drug effect was described by an inhibitory sigmoidal Emax model. Since TG levels change with the introduction of a meal, a bi-exponential meal effect model was utilized. The total change in TG was fitted using a turnover model with drug and meal effects on the TG production rate. The current analysis presents a PKPD modeling strategy of time-varying TG data coming from both endogenous and exogenous sources. In general, the presented model could be utilized in the model-based drug development of drugs that influence TG levels in blood.
Collapse
Affiliation(s)
- Malek Okour
- Clinical Pharmacology Modeling and Simulation (CPMS), GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426-0989, USA
| | - Richard A Brigandi
- Exploratory Discovery, GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426-0989, USA
| | - David Tenero
- Clinical Pharmacology Modeling and Simulation (CPMS), GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA, 19426-0989, USA
| |
Collapse
|
24
|
Hernández-Corbacho MJ, Obeid LM. A novel role for DGATs in cancer. Adv Biol Regul 2018; 72:89-101. [PMID: 30579761 DOI: 10.1016/j.jbior.2018.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/09/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023]
Affiliation(s)
- María José Hernández-Corbacho
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY, 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY, 11794, USA; The Northport VA Medical Center, Northport, NY, 11768, USA.
| |
Collapse
|
25
|
Huang LS, Yuen JJ, Trites MJ, Saha A, Epps CT, Hu Y, Kerolle S, Lee SA, Jiang H, Goldberg IJ, Blaner WS, Clugston RD. Dietary Macronutrient Composition Determines the Contribution of DGAT1 to Alcoholic Steatosis. Alcohol Clin Exp Res 2018; 42:2298-2312. [PMID: 30192394 PMCID: PMC6286229 DOI: 10.1111/acer.13881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/02/2018] [Indexed: 01/01/2023]
Abstract
Background The first stage of alcoholic liver disease is hepatic steatosis. While alcohol is known to profoundly impact hepatic lipid metabolism, gaps in our knowledge remain regarding the mechanisms leading to alcohol‐induced hepatic triglyceride (TG) accumulation. As the sole enzymes catalyzing the final step in TG synthesis, diacylglycerol O‐acyltransferase (DGAT) 1 and 2 are potentially important contributors to alcoholic steatosis. Our goal was to study the effects of dietary fat content on alcohol‐induced hepatic TG accumulation, and the relative contribution of DGAT1 and DGAT2 to alcoholic steatosis. Methods These studies were carried out in wild‐type (WT) mice fed alcohol‐containing high‐fat or low‐fat formulations of Lieber‐DeCarli liquid diets, as well as follow‐up studies in Dgat1−/− mice. Results A direct comparison of the low‐fat and high‐fat liquid diet in WT mice revealed surprisingly similar levels of alcoholic steatosis, although there were underlying differences in the pattern of hepatic lipid accumulation and expression of genes involved in hepatic lipid metabolism. Follow‐up studies in Dgat1−/− mice revealed that these animals are protected from alcoholic steatosis when consumed as part of a high‐fat diet, but not a low‐fat diet. Conclusions Dietary macronutrient composition influences the relative contribution of DGAT1 and DGAT2 to alcoholic steatosis, such that in the context of alcohol and a high‐fat diet, DGAT1 predominates.
Collapse
Affiliation(s)
- Li-Shin Huang
- Department of Medicine, Columbia University, New York, New York
| | - Jason J Yuen
- Department of Medicine, Columbia University, New York, New York
| | - Michael J Trites
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Amit Saha
- Department of Medicine, Columbia University, New York, New York
| | - Caleb T Epps
- Department of Medicine, Columbia University, New York, New York
| | - Yungying Hu
- Department of Medicine, New York University Langone Medical Center, New York, New York
| | | | - Seung-Ah Lee
- Department of Medicine, Columbia University, New York, New York
| | - Hongfeng Jiang
- Department of Medicine, Columbia University, New York, New York
| | - Ira J Goldberg
- Department of Medicine, New York University Langone Medical Center, New York, New York
| | | | - Robin D Clugston
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
26
|
van Rijn JM, Ardy RC, Kuloğlu Z, Härter B, van Haaften-Visser DY, van der Doef HP, van Hoesel M, Kansu A, van Vugt AH, Thian M, Kokke FT, Krolo A, Başaran MK, Kaya NG, Aksu AÜ, Dalgıç B, Ozcay F, Baris Z, Kain R, Stigter EC, Lichtenbelt KD, Massink MP, Duran KJ, Verheij JB, Lugtenberg D, Nikkels PG, Brouwer HG, Verkade HJ, Scheenstra R, Spee B, Nieuwenhuis EE, Coffer PJ, Janecke AR, van Haaften G, Houwen RH, Müller T, Middendorp S, Boztug K. Intestinal Failure and Aberrant Lipid Metabolism in Patients With DGAT1 Deficiency. Gastroenterology 2018; 155:130-143.e15. [PMID: 29604290 PMCID: PMC6058035 DOI: 10.1053/j.gastro.2018.03.040] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/19/2018] [Accepted: 03/22/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Congenital diarrheal disorders are rare inherited intestinal disorders characterized by intractable, sometimes life-threatening, diarrhea and nutrient malabsorption; some have been associated with mutations in diacylglycerol-acyltransferase 1 (DGAT1), which catalyzes formation of triacylglycerol from diacylglycerol and acyl-CoA. We investigated the mechanisms by which DGAT1 deficiency contributes to intestinal failure using patient-derived organoids. METHODS We collected blood samples from 10 patients, from 6 unrelated pedigrees, who presented with early-onset severe diarrhea and/or vomiting, hypoalbuminemia, and/or (fatal) protein-losing enteropathy with intestinal failure; we performed next-generation sequencing analysis of DNA from 8 patients. Organoids were generated from duodenal biopsies from 3 patients and 3 healthy individuals (controls). Caco-2 cells and patient-derived dermal fibroblasts were transfected or transduced with vectors that express full-length or mutant forms of DGAT1 or full-length DGAT2. We performed CRISPR/Cas9-guided disruption of DGAT1 in control intestinal organoids. Cells and organoids were analyzed by immunoblot, immunofluorescence, flow cytometry, chromatography, quantitative real-time polymerase chain reaction, and for the activity of caspases 3 and 7. RESULTS In the 10 patients, we identified 5 bi-allelic loss-of-function mutations in DGAT1. In patient-derived fibroblasts and organoids, the mutations reduced expression of DGAT1 protein and altered triacylglycerol metabolism, resulting in decreased lipid droplet formation after oleic acid addition. Expression of full-length DGAT2 in patient-derived fibroblasts restored formation of lipid droplets. Organoids derived from patients with DGAT1 mutations were more susceptible to lipid-induced cell death than control organoids. CONCLUSIONS We identified a large cohort of patients with congenital diarrheal disorders with mutations in DGAT1 that reduced expression of its product; dermal fibroblasts and intestinal organoids derived from these patients had altered lipid metabolism and were susceptible to lipid-induced cell death. Expression of full-length wildtype DGAT1 or DGAT2 restored normal lipid metabolism in these cells. These findings indicate the importance of DGAT1 in fat metabolism and lipotoxicity in the intestinal epithelium. A fat-free diet might serve as the first line of therapy for patients with reduced DGAT1 expression. It is important to identify genetic variants associated with congenital diarrheal disorders for proper diagnosis and selection of treatment strategies.
Collapse
Affiliation(s)
- Jorik M. van Rijn
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht University, Utrecht, The Netherlands,Regenerative Medicine Center, Utrecht University, Utrecht, The Netherlands
| | - Rico Chandra Ardy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Zarife Kuloğlu
- Department of Pediatric Gastroenterology, Ankara University School of Medicine, Ankara, Turkey
| | - Bettina Härter
- Division of Paediatric Surgery, Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Innsbruck, Austria
| | - Désirée Y. van Haaften-Visser
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht University, Utrecht, The Netherlands,Regenerative Medicine Center, Utrecht University, Utrecht, The Netherlands
| | - Hubert P.J. van der Doef
- Department of Pediatric Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marliek van Hoesel
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht University, Utrecht, The Netherlands,Regenerative Medicine Center, Utrecht University, Utrecht, The Netherlands
| | - Aydan Kansu
- Department of Pediatric Gastroenterology, Ankara University School of Medicine, Ankara, Turkey
| | - Anke H.M. van Vugt
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht University, Utrecht, The Netherlands,Regenerative Medicine Center, Utrecht University, Utrecht, The Netherlands
| | - Marini Thian
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Freddy T.M. Kokke
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht University, Utrecht, The Netherlands
| | - Ana Krolo
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Meryem Keçeli Başaran
- Pediatric Gastroenterology Department, Akdeniz University Medicine Hospital, Antalya, Turkey
| | - Neslihan Gurcan Kaya
- Department of Pediatric Gastroenterology, Gazi University School of Medicine, Ankara, Turkey
| | - Aysel Ünlüsoy Aksu
- Department of Pediatric Gastroenterology, Gazi University School of Medicine, Ankara, Turkey
| | - Buket Dalgıç
- Department of Pediatric Gastroenterology, Gazi University School of Medicine, Ankara, Turkey
| | - Figen Ozcay
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Faculty of Medicine, Başkent University, Ankara, Turkey
| | - Zeren Baris
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Faculty of Medicine, Başkent University, Ankara, Turkey
| | - Renate Kain
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Edwin C.A. Stigter
- Molecular Cancer Research, Center Molecular Medicine, Utrecht University, Utrecht, The Netherlands
| | - Klaske D. Lichtenbelt
- Department of Medical Genetics, Center for Molecular Medicine, Utrecht University, Utrecht, The Netherlands
| | - Maarten P.G. Massink
- Department of Medical Genetics, Center for Molecular Medicine, Utrecht University, Utrecht, The Netherlands
| | - Karen J. Duran
- Department of Medical Genetics, Center for Molecular Medicine, Utrecht University, Utrecht, The Netherlands
| | - Joke B.G.M Verheij
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dorien Lugtenberg
- Department of Human Genetics, Radboud University Nijmegen Medical Center, Nijmegen The Netherlands
| | - Peter G.J. Nikkels
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Henkjan J. Verkade
- Department of Pediatric Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, The Netherlands
| | - René Scheenstra
- Department of Pediatric Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands
| | - Edward E.S. Nieuwenhuis
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht University, Utrecht, The Netherlands
| | - Paul J. Coffer
- Regenerative Medicine Center, Utrecht University, Utrecht, The Netherlands
| | - Andreas R. Janecke
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Gijs van Haaften
- Department of Medical Genetics, Center for Molecular Medicine, Utrecht University, Utrecht, The Netherlands
| | - Roderick H.J. Houwen
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children’s Hospital, Utrecht University, Utrecht, The Netherlands
| | - Thomas Müller
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Sabine Middendorp
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands; Regenerative Medicine Center, Utrecht University, Utrecht, The Netherlands.
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria; St. Anna Kinderspital and Children's Cancer Research Institute, Department of Pediatrics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
27
|
Xiao C, Stahel P, Carreiro AL, Buhman KK, Lewis GF. Recent Advances in Triacylglycerol Mobilization by the Gut. Trends Endocrinol Metab 2018; 29:151-163. [PMID: 29306629 DOI: 10.1016/j.tem.2017.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 11/26/2022]
Abstract
Dietary lipid absorption and lipoprotein secretion by the gut are important in maintaining whole-body energy homeostasis and have significant implications for health and disease. The processing of dietary lipids, including storage within and subsequent mobilization and transport from enterocyte cytoplasmic lipid droplets or other intestinal lipid storage pools (including the secretary pathway, lamina propria and lymphatics) and secretion of chylomicrons, involves coordinated steps that are subject to various controls. This review summarizes recent advances in our understanding of the mechanisms that underlie lipid storage and mobilization by small intestinal enterocytes and the intestinal lymphatic vasculature. Therapeutic targeting of lipid processing by the gut may provide opportunities for the treatment and prevention of dyslipidemia, and for improving health status.
Collapse
Affiliation(s)
- Changting Xiao
- Departments of Medicine and Physiology, Division of Endocrinology and Metabolism, Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Priska Stahel
- Departments of Medicine and Physiology, Division of Endocrinology and Metabolism, Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Alicia L Carreiro
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Gary F Lewis
- Departments of Medicine and Physiology, Division of Endocrinology and Metabolism, Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Hung YH, Carreiro AL, Buhman KK. Dgat1 and Dgat2 regulate enterocyte triacylglycerol distribution and alter proteins associated with cytoplasmic lipid droplets in response to dietary fat. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:600-614. [PMID: 28249764 PMCID: PMC5503214 DOI: 10.1016/j.bbalip.2017.02.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/31/2017] [Accepted: 02/24/2017] [Indexed: 12/16/2022]
Abstract
Enterocytes, the absorptive cells of the small intestine, mediate efficient absorption of dietary fat (triacylglycerol, TAG). The digestive products of dietary fat are taken up by enterocytes, re-esterified into TAG, and packaged on chylomicrons (CMs) for secretion into blood or temporarily stored within cytoplasmic lipid droplets (CLDs). Altered enterocyte TAG distribution impacts susceptibility to high fat diet associated diseases, but molecular mechanisms directing TAG toward these fates are unclear. Two enzymes, acyl CoA: diacylglycerol acyltransferase 1 (Dgat1) and Dgat2, catalyze the final, committed step of TAG synthesis within enterocytes. Mice with intestine-specific overexpression of Dgat1 (Dgat1Int) or Dgat2 (Dgat2Int), or lack of Dgat1 (Dgat1-/-), were previously found to have altered intestinal TAG secretion and storage. We hypothesized that varying intestinal Dgat1 and Dgat2 levels alters TAG distribution in subcellular pools for CM synthesis as well as the morphology and proteome of CLDs. To test this we used ultrastructural and proteomic methods to investigate intracellular TAG distribution and CLD-associated proteins in enterocytes from Dgat1Int, Dgat2Int, and Dgat1-/- mice 2h after a 200μl oral olive oil gavage. We found that varying levels of intestinal Dgat1 and Dgat2 altered TAG pools involved in CM assembly and secretion, the number or size of CLDs present in enterocytes, and the enterocyte CLD proteome. Overall, these results support a model where Dgat1 and Dgat2 function coordinately to regulate the process of dietary fat absorption by preferentially synthesizing TAG for incorporation into distinct subcellular TAG pools in enterocytes.
Collapse
Affiliation(s)
- Yu-Han Hung
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Alicia L Carreiro
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
29
|
Gane E, Stedman C, Dole K, Chen J, Meyers CD, Wiedmann B, Zhang J, Raman P, Colvin RA. A Diacylglycerol Transferase 1 Inhibitor Is a Potent Hepatitis C Antiviral in Vitro but Not in Patients in a Randomized Clinical Trial. ACS Infect Dis 2017; 3:144-151. [PMID: 27788579 DOI: 10.1021/acsinfecdis.6b00138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hepatitis C virus (HCV) infection is a significant cause of liver disease affecting 80-150 million people globally. Diacylglycerol transferase 1 (DGAT-1), a triglyceride synthesis enzyme, is important for the HCV life cycle in vitro. Pradigastat, a potent DGAT-1 inhibitor found to lower triglycerides and HgbA1c in patients, was investigated for safety and efficacy in patients with HCV. This was a two-part study. In the in vitro study, the effect of pradigastat on virus production was evaluated in infected cells in culture. In the clinical study ( https://clinicaltrials.gov/ct2/show/NCT01387958 ), 32 patients with HCV infection were randomized to receive pradigastat or placebo (26:6) once daily for 14 days. Primary efficacy outcomes were serum viral RNA and alanine aminotransferase levels. In vitro, pradigastat significantly reduced virus production, consistent with inhibition of viral assembly and release. However, the clinical study was prematurely terminated for lack of efficacy. There was no significant change in serum viral RNA levels after dosing with pradigastat or placebo for 14 days. Pradigastat was safe and well-tolerated in this population. Most treatment-emergent adverse events were gastrointestinal; there were no hepatic adverse events. Although pradigastat had a potent antiviral effect in vitro, no significant antiviral effect was observed in patients at predicted efficacious exposures.
Collapse
Affiliation(s)
- Edward Gane
- Auckland Clinical Sciences , Grafton, Auckland 1010, New Zealand
| | - Catherine Stedman
- Christchurch Hospital and University of Otago , Christchurch 4710, New Zealand
| | - Kiran Dole
- Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Jin Chen
- Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Charles Daniel Meyers
- Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Brigitte Wiedmann
- Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Jin Zhang
- Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Prakash Raman
- Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Richard A Colvin
- Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| |
Collapse
|
30
|
Upthagrove A, Chen J, Meyers CD, Kulmatycki K, Bretz A, Wang L, Peng L, Palamar S, Lin M, Majumdar T, Tran P, Einolf HJ. Pradigastat disposition in humans: in vivo and in vitro investigations. Xenobiotica 2016; 47:1077-1089. [PMID: 27855567 DOI: 10.1080/00498254.2016.1263405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
1. Pradigastat is a potent and specific diacylglycerol acyltransferase-1 (DGAT1) inhibitor effective in lowering postprandial triglycerides (TG) in healthy human subjects and fasting TG in familial chylomicronemia syndrome (FCS) patients. 2. Here we present the results of human oral absorption, metabolism and excretion (AME), intravenous pharmacokinetic (PK), and in vitro studies which together provide an overall understanding of the disposition of pradigastat in humans. 3. In human in vitro systems, pradigastat is metabolized slowly to a stable acyl glucuronide (M18.4), catalyzed mainly by UDP-glucuronosyltransferases (UGT) 1A1, UGT1A3 and UGT2B7. M18.4 was observed at very low levels in human plasma. 4. In the human AME study, pradigastat was recovered in the feces as parent drug, confounding the assessment of pradigastat absorption and the important routes of elimination. However, considering pradigastat exposure after oral and intravenous dosing, this data suggests that pradigastat was completely bioavailable in the radiolabeled AME study and therefore completely absorbed. 5. Pradigastat is eliminated very slowly into the feces, presumably via the bile. Renal excretion is negligible. Oxidative metabolism is minimal. The extent to which pradigastat is eliminated via metabolism to M18.4 could not be established from these studies due to the inherent instability of glucuronides in the gastrointestinal tract.
Collapse
Affiliation(s)
- Alana Upthagrove
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| | - Jin Chen
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| | - Charles D Meyers
- b Translational Medicine, Novartis Institutes for Biomedical Research , Cambridge , MA , USA
| | - Kenneth Kulmatycki
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| | - Angela Bretz
- c The Genomics Institute of the Novartis Research Foundation, Novartis Institutes for Biomedical Research , San Diego , CA , USA , and
| | - Lai Wang
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| | - Lana Peng
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| | - Safet Palamar
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| | - Melissa Lin
- d Technical Research and Development, Novartis Pharmaceuticals , East Hanover , NJ , USA
| | - Tapan Majumdar
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| | - Phi Tran
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| | - Heidi J Einolf
- a Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , NJ , USA
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW We provide an overview of orally administered lipid-lowering therapies under development. RECENT FINDINGS Recent data support statins for intermediate risk primary prevention, and ezetimibe for high-risk secondary prevention. Novel agents in development include bempedoic acid and gemcabene, and work continues on one remaining cholesteryl ester transfer protein inhibitor, anacetrapib, to determine whether this class can reduce cardiovascular risk. Selective peroxisome proliferator-activated receptor modulators such as K-877 are under study to determine whether they have an advantage over older fibrates. Diacylglycerol transferase inhibitors such as pradigastat appear to have potent triglyceride-lowering effects, even for patients with familial chylomicronemia syndrome. Finally, novel ω-3 preparations are available with significant triglyceride lowering, although their role in therapy remains unclear. SUMMARY Statins will remain the backbone of lipid-lowering therapy, although several novel oral agents are promising. The common theme across drugs in development is the demonstration of good lipid-lowering effect, although lacking cardiovascular outcomes data, which will likely be necessary before any of them, can be recommended or approved for widespread use.
Collapse
Affiliation(s)
- Steven E Gryn
- Department of Medicine, Schulich School of Medicine and Dentistry, London Health Sciences Centre-University Hospital, Western University, London, Ontario, Canada
| | | |
Collapse
|
32
|
Okopień B, Bułdak Ł, Bołdys A. Current and future trends in the lipid lowering therapy. Pharmacol Rep 2016; 68:737-47. [DOI: 10.1016/j.pharep.2016.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/25/2016] [Accepted: 03/25/2016] [Indexed: 12/31/2022]
|
33
|
Meyers CD, Noe A, Salunke A, Movva A, Kulmatycki K, Neelakantham S, Crissey A, Majumdar T, Chen J. Effect of Pradigastat, a Diacylglycerol Acyltransferase 1 Inhibitor, on the QTcF Interval in Humans. Clin Pharmacol Drug Dev 2016; 5:450-459. [DOI: 10.1002/cpdd.278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 05/11/2016] [Accepted: 05/23/2016] [Indexed: 11/11/2022]
Affiliation(s)
| | - Adele Noe
- Novartis Pharma AG; Basel Switzerland
| | - Atish Salunke
- Novartis Healthcare Private Limited; Hyderabad Telangana India
| | - Aishwarya Movva
- Novartis Institutes for BioMedical Research; Cambridge MA USA
| | | | | | - Anne Crissey
- Novartis Institutes for BioMedical Research; Cambridge MA USA
| | - Tapan Majumdar
- Novartis Institutes for BioMedical Research; East Hanover NJ USA
| | - Jin Chen
- Novartis Institutes for BioMedical Research; East Hanover NJ USA
| |
Collapse
|
34
|
Xiao C, Dash S, Morgantini C, Hegele RA, Lewis GF. Pharmacological Targeting of the Atherogenic Dyslipidemia Complex: The Next Frontier in CVD Prevention Beyond Lowering LDL Cholesterol. Diabetes 2016; 65:1767-78. [PMID: 27329952 DOI: 10.2337/db16-0046] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 03/23/2016] [Indexed: 11/13/2022]
Abstract
Notwithstanding the effectiveness of lowering LDL cholesterol, residual CVD risk remains in high-risk populations, including patients with diabetes, likely contributed to by non-LDL lipid abnormalities. In this Perspectives in Diabetes article, we emphasize that changing demographics and lifestyles over the past few decades have resulted in an epidemic of the "atherogenic dyslipidemia complex," the main features of which include hypertriglyceridemia, low HDL cholesterol levels, qualitative changes in LDL particles, accumulation of remnant lipoproteins, and postprandial hyperlipidemia. We briefly review the underlying pathophysiology of this form of dyslipidemia, in particular its association with insulin resistance, obesity, and type 2 diabetes, and the marked atherogenicity of this condition. We explain the failure of existing classes of therapeutic agents such as fibrates, niacin, and cholesteryl ester transfer protein inhibitors that are known to modify components of the atherogenic dyslipidemia complex. Finally, we discuss targeted repurposing of existing therapies and review promising new therapeutic strategies to modify the atherogenic dyslipidemia complex. We postulate that targeting the central abnormality of the atherogenic dyslipidemia complex, the elevation of triglyceride-rich lipoprotein particles, represents a new frontier in CVD prevention and is likely to prove the most effective strategy in correcting most aspects of the atherogenic dyslipidemia complex, thereby preventing CVD events.
Collapse
Affiliation(s)
- Changting Xiao
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Satya Dash
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Cecilia Morgantini
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Robert A Hegele
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Gary F Lewis
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
35
|
D'Aquila T, Hung YH, Carreiro A, Buhman KK. Recent discoveries on absorption of dietary fat: Presence, synthesis, and metabolism of cytoplasmic lipid droplets within enterocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:730-47. [PMID: 27108063 DOI: 10.1016/j.bbalip.2016.04.012] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/16/2016] [Accepted: 04/16/2016] [Indexed: 02/07/2023]
Abstract
Dietary fat provides essential nutrients, contributes to energy balance, and regulates blood lipid concentrations. These functions are important to health, but can also become dysregulated and contribute to diseases such as obesity, diabetes, cardiovascular disease, and cancer. Within enterocytes, the digestive products of dietary fat are re-synthesized into triacylglycerol, which is either secreted on chylomicrons or stored within cytoplasmic lipid droplets (CLDs). CLDs were originally thought to be inert stores of neutral lipids, but are now recognized as dynamic organelles that function in multiple cellular processes in addition to lipid metabolism. This review will highlight recent discoveries related to dietary fat absorption with an emphasis on the presence, synthesis, and metabolism of CLDs within this process.
Collapse
Affiliation(s)
- Theresa D'Aquila
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Yu-Han Hung
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Alicia Carreiro
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
36
|
Mendonza A, Hanna I, Meyers D, Koo P, Neelakantham S, Zhu B, Majumdar T, Rebello S, Sunkara G, Chen J. Assessment of pharmacokinetic drug-drug interaction between pradigastat and atazanavir or probenecid. J Clin Pharmacol 2015; 56:355-64. [PMID: 26189431 DOI: 10.1002/jcph.595] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/15/2015] [Indexed: 11/08/2022]
Abstract
Pradigastat, a novel diacylglycerol acyltransferase-1 inhibitor, has activity in common metabolic diseases associated with abnormal accumulation of triglycerides. In vitro studies suggest that glucuronidation is the predominant metabolism pathway for elimination of pradigastat in humans and confirmed the role of uridine 5'-diphosphoglucuronosyltransferase (UGT) enzymes, UGT1A1, -1A3, and -2B7. The in vitro studies using atazanavir as a selective inhibitor of UGT1A1 and -1A3 indicated that these enzymes contribute ∼55% toward the overall glucuronidation pathway. Therefore, a clinical study was conducted to assess the potential for drug interaction between pradigastat and probenecid (purported general UGT inhibitor) or atazanavir (selective UGT1A1, -1A3 inhibitor). The study included 2 parallel cohorts, each with 3 sequential treatment periods and 22 healthy subjects per cohort. The 90%CI of the geometric mean ratios for Cmax,ss and AUCτ,ss of pradigastat were within 0.80-1.25 when administered in combination with probenecid. However, the Cmax,ss and AUCτ,ss of pradigastat decreased by 31% (90%CI: 0.62-0.78) and 26% (0.67-0.82), respectively, when administered in combination with atazanavir. This magnitude of decrease in pradigastat steady-state exposure is not considered clinically relevant. Pradigastat was well tolerated by all subjects, either alone or in combination with atazanavir or probenecid.
Collapse
Affiliation(s)
- Anisha Mendonza
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Imad Hanna
- Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
| | - Dan Meyers
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Phillip Koo
- Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
| | | | - Bing Zhu
- Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
| | | | - Sam Rebello
- Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
| | | | - Jin Chen
- Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
| |
Collapse
|