1
|
Wilkerson JL, Tatum SM, Holland WL, Summers SA. Ceramides are fuel gauges on the drive to cardiometabolic disease. Physiol Rev 2024; 104:1061-1119. [PMID: 38300524 PMCID: PMC11381030 DOI: 10.1152/physrev.00008.2023] [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: 02/14/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/02/2024] Open
Abstract
Ceramides are signals of fatty acid excess that accumulate when a cell's energetic needs have been met and its nutrient storage has reached capacity. As these sphingolipids accrue, they alter the metabolism and survival of cells throughout the body including in the heart, liver, blood vessels, skeletal muscle, brain, and kidney. These ceramide actions elicit the tissue dysfunction that underlies cardiometabolic diseases such as diabetes, coronary artery disease, metabolic-associated steatohepatitis, and heart failure. Here, we review the biosynthesis and degradation pathways that maintain ceramide levels in normal physiology and discuss how the loss of ceramide homeostasis drives cardiometabolic pathologies. We highlight signaling nodes that sense small changes in ceramides and in turn reprogram cellular metabolism and stimulate apoptosis. Finally, we evaluate the emerging therapeutic utility of these unique lipids as biomarkers that forecast disease risk and as targets of ceramide-lowering interventions that ameliorate disease.
Collapse
Affiliation(s)
- Joseph L Wilkerson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
2
|
Hu K, Zhang Q, Chen Y, Yang J, Xia Y, Rao B, Li S, Shen Y, Cao M, Lu H, Qin A, Jiang XC, Yao D, Zhao J, Zhou L, Cao Y. Cryo-EM structure of human sphingomyelin synthase and its mechanistic implications for sphingomyelin synthesis. Nat Struct Mol Biol 2024; 31:884-895. [PMID: 38388831 DOI: 10.1038/s41594-024-01237-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 01/31/2024] [Indexed: 02/24/2024]
Abstract
Sphingomyelin (SM) has key roles in modulating mammalian membrane properties and serves as an important pool for bioactive molecules. SM biosynthesis is mediated by the sphingomyelin synthase (SMS) family, comprising SMS1, SMS2 and SMS-related (SMSr) members. Although SMS1 and SMS2 exhibit SMS activity, SMSr possesses ceramide phosphoethanolamine synthase activity. Here we determined the cryo-electron microscopic structures of human SMSr in complexes with ceramide, diacylglycerol/phosphoethanolamine and ceramide/phosphoethanolamine (CPE). The structures revealed a hexameric arrangement with a reaction chamber located between the transmembrane helices. Within this structure, a catalytic pentad E-H/D-H-D was identified, situated at the interface between the lipophilic and hydrophilic segments of the reaction chamber. Additionally, the study unveiled the two-step synthesis process catalyzed by SMSr, involving PE-PLC (phosphatidylethanolamine-phospholipase C) hydrolysis and the subsequent transfer of the phosphoethanolamine moiety to ceramide. This research provides insights into the catalytic mechanism of SMSr and expands our understanding of sphingolipid metabolism.
Collapse
Affiliation(s)
- Kexin Hu
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Zhang
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yang Chen
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jintong Yang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Ying Xia
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Rao
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaobai Li
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafeng Shen
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mi Cao
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongliang Lu
- College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Deqiang Yao
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Aging & Tissue Regeneration, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jie Zhao
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lu Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China.
| | - Yu Cao
- Institute of Precision Medicine, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Pihlström S, Richardt S, Määttä K, Pekkinen M, Olkkonen VM, Mäkitie O, Mäkitie RE. SGMS2 in primary osteoporosis with facial nerve palsy. Front Endocrinol (Lausanne) 2023; 14:1224318. [PMID: 37886644 PMCID: PMC10598846 DOI: 10.3389/fendo.2023.1224318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023] Open
Abstract
Pathogenic heterozygous variants in SGMS2 cause a rare monogenic form of osteoporosis known as calvarial doughnut lesions with bone fragility (CDL). The clinical presentations of SGMS2-related bone pathology range from childhood-onset osteoporosis with low bone mineral density and sclerotic doughnut-shaped lesions in the skull to a severe spondylometaphyseal dysplasia with neonatal fractures, long-bone deformities, and short stature. In addition, neurological manifestations occur in some patients. SGMS2 encodes sphingomyelin synthase 2 (SMS2), an enzyme involved in the production of sphingomyelin (SM). This review describes the biochemical structure of SM, SM metabolism, and their molecular actions in skeletal and neural tissue. We postulate how disrupted SM gradient can influence bone formation and how animal models may facilitate a better understanding of SGMS2-related osteoporosis.
Collapse
Affiliation(s)
- Sandra Pihlström
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sampo Richardt
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi Määttä
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Minna Pekkinen
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children´s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Vesa M. Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Outi Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children´s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Riikka E. Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Otorhinolaryngology – Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Li Z, He M, Chen G, Souaiaia T, Worgall TS, Jiang XC. Effect of Total SMS Activity on LDL Catabolism in Mice. Arterioscler Thromb Vasc Biol 2023; 43:1251-1261. [PMID: 37128925 PMCID: PMC10330209 DOI: 10.1161/atvbaha.123.319031] [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: 02/02/2023] [Accepted: 04/06/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Sphingomyelin (SM) and cholesterol are 2 key lipid partners on cell membranes and on lipoproteins. Many studies have indicated the influence of cholesterol on SM metabolism. This study examined the influence of SM biosynthesis on cholesterol metabolism. METHODS Inducible global Sms1 KO (knockout)/global Sms2 KO mice were prepared to evaluate the effect of whole-body SM biosynthesis deficiency on lipoprotein metabolism. Tissue cholesterol, SM, ceramide, and glucosylceramide levels were measured. Triglyceride production rate and LDL (low-density lipoprotein) catabolism were measured. Lipid rafts were isolated and LDL receptor mass and function were evaluated. Also, the effects of exogenous sphingolipids on hepatocytes were investigated. RESULTS We found that total SMS (SM synthase) depletion significantly reduced plasma SM levels. Also, the total deficiency significantly induced plasma cholesterol, apoB (apolipoprotein B), and apoE (apolipoprotein E) levels. Importantly, total SMS deficiency, but not SMS2 deficiency, dramatically decreased LDL receptors in the liver and attenuated LDL uptake through the receptor. Further, we found that total SMS deficiency greatly reduced LDL receptors in the lipid rafts, which contained significantly lower SM and significantly higher glucosylceramide, as well as cholesterol. Furthermore, we treated primary hepatocytes and Huh7 cells (a human hepatoma cell line) with SM, ceramide, or glucosylceramide, and we found that only SM could upregulate LDL receptor levels in a dose-dependent fashion. CONCLUSIONS Whole-body SM biosynthesis plays an important role in LDL cholesterol catabolism. The total SMS deficiency, but not SMS2 deficiency, reduces LDL uptake and causes LDL cholesterol accumulation in the circulation. Given the fact that serum SM level is a risk factor for cardiovascular diseases, inhibiting SMS2 but not SMS1 should be the desirable approach.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn (Z.L., M.H., G.C., T.S., X.-C.J.)
- Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System (Z.L., X.-C.J.)
| | - Mulin He
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn (Z.L., M.H., G.C., T.S., X.-C.J.)
| | - Guangzhi Chen
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn (Z.L., M.H., G.C., T.S., X.-C.J.)
| | - Tade Souaiaia
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn (Z.L., M.H., G.C., T.S., X.-C.J.)
| | - Tilla S Worgall
- Department of Pathology and Cell Biology, Columbia University, New York (T.S.W.)
| | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn (Z.L., M.H., G.C., T.S., X.-C.J.)
- Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System (Z.L., X.-C.J.)
| |
Collapse
|
5
|
Sojo L, Santos-González E, Riera L, Aguilera A, Barahona R, Pellicer P, Buxó M, Mayneris-Perxachs J, Fernandez-Balsells M, Fernández-Real JM. Plasma Lipidomics Profiles Highlight the Associations of the Dual Antioxidant/Pro-oxidant Molecules Sphingomyelin and Phosphatidylcholine with Subclinical Atherosclerosis in Patients with Type 1 Diabetes. Antioxidants (Basel) 2023; 12:antiox12051132. [PMID: 37237999 DOI: 10.3390/antiox12051132] [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: 04/07/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Here, we report on our study of plasma lipidomics profiles of patients with type 1 diabetes (T1DM) and explore potential associations. One hundred and seven patients with T1DM were consecutively recruited. Ultrasound imaging of peripheral arteries was performed using a high image resolution B-mode ultrasound system. Untargeted lipidomics analysis was performed using UHPLC coupled to qTOF/MS. The associations were evaluated using machine learning algorithms. SM(32:2) and ether lipid species (PC(O-30:1)/PC(P-30:0)) were significantly and positively associated with subclinical atherosclerosis (SA). This association was further confirmed in patients with overweight/obesity (specifically with SM(40:2)). A negative association between SA and lysophosphatidylcholine species was found among lean subjects. Phosphatidylcholines (PC(40:6) and PC(36:6)) and cholesterol esters (ChoE(20:5)) were associated positively with intima-media thickness both in subjects with and without overweight/obesity. In summary, the plasma antioxidant molecules SM and PC differed according to the presence of SA and/or overweight status in patients with T1DM. This is the first study showing the associations in T1DM, and the findings may be useful in the targeting of a personalized approach aimed at preventing cardiovascular disease in these patients.
Collapse
Affiliation(s)
- Lidia Sojo
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17007 Girona, Spain
| | - Elena Santos-González
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17007 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Lídia Riera
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
| | - Alex Aguilera
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, 17003 Girona, Spain
| | - Rebeca Barahona
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, 17003 Girona, Spain
| | - Paula Pellicer
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
| | - Maria Buxó
- Girona Biomedical Research Institute (IDIBGI), 17007 Girona, Spain
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17007 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Mercè Fernandez-Balsells
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17007 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, 17003 Girona, Spain
| | - José-Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17007 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, 17003 Girona, Spain
| |
Collapse
|
6
|
Effect of Total Sphingomyelin Synthase Activity on Low Density Lipoprotein Catabolism in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.527088. [PMID: 36798262 PMCID: PMC9934588 DOI: 10.1101/2023.02.03.527088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Background Sphingomyelin (SM) and cholesterol are two key lipid partners on cell membranes and on lipoproteins. Many studies have indicated the influence of cholesterol on SM metabolism. This study examined the influence of SM biosynthesis on cholesterol metabolism. Methods Inducible global Sms1 KO/global Sms2 KO mice were prepared to evaluate the effect of whole-body SM biosynthesis deficiency on lipoprotein metabolism. Tissue cholesterol, SM, ceramide, and glucosylceramide levels were measured. TG production rate and LDL catabolism were measured. Lipid rafts were isolated and LDL receptor mass and function were evaluated. Also, the effects of exogenous sphingolipids on hepatocytes were investigated. Results We found that total SMS depletion significantly reduced plasma SM levels. Also, the total deficiency significantly induced plasma cholesterol, apoB, and apoE levels. Importantly, total SMS deficiency, but not SMS2 deficiency, dramatically decreased LDL receptors in the liver and attenuated LDL uptake through the receptor. Further, we found that total SMS deficiency greatly reduced LDL receptors in the lipid rafts which contained significantly lower SM and significantly higher glucosylceramide as well as cholesterol. Furthermore, we treated primary hepatocytes and Huh7 cells (a human hepatoma cell line) with SM, ceramide, or glucosylceramide, and we found that only SM could up-regulate LDL receptor levels in a dose-dependent fashion. Conclusions Whole-body SM biosynthesis plays an important role in LDL-cholesterol catabolism. The total SMS deficiency, but not SMS2 deficiency, reduces LDL uptake and causes LDL-cholesterol accumulation in the circulation. Given the fact that serum SM level is a risk factor for cardiovascular diseases, inhibiting SMS2 but not SMS1 should be the desirable approach. Graphic Abstract
Collapse
|
7
|
Yang F, Chen G. The nutritional functions of dietary sphingomyelin and its applications in food. Front Nutr 2022; 9:1002574. [PMID: 36337644 PMCID: PMC9626766 DOI: 10.3389/fnut.2022.1002574] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Sphingolipids are common structural components of cell membranes and are crucial for cell functions in physiological and pathophysiological conditions. Sphingomyelin and its metabolites, such as sphingoid bases, ceramide, ceramide-1-phosphate, and sphingosine-1-phosphate, play signaling roles in the regulation of human health. The diverse structures of sphingolipids elicit various functions in cellular membranes and signal transduction, which may affect cell growth, differentiation, apoptosis, and maintain biological activities. As nutrients, dietary sphingomyelin and its metabolites have wide applications in the food and pharmaceutical industry. In this review, we summarized the distribution, classifications, structures, digestion, absorption and metabolic pathways of sphingolipids, and discussed the nutritional functioning of sphingomyelin in chronic metabolic diseases. The possible implications of dietary sphingomyelin in the modern food preparations including dairy products and infant formula, skin improvement, delivery system and oil organogels are also evaluated. The production of endogenous sphingomyelin is linked to pathological changes in obesity, diabetes, and atherosclerosis. However, dietary supplementations of sphingomyelin and its metabolites have been shown to maintain cholesterol homeostasis and lipid metabolism, and to prevent or treat these diseases. This seemly paradoxical phenomenon shows that dietary sphingomyelin and its metabolites are candidates for food additives and functional food development for the prevention and treatment of chronic metabolic diseases in humans.
Collapse
Affiliation(s)
- Fang Yang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
- *Correspondence: Fang Yang,
| | - Guoxun Chen
- Department of Nutrition, The University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
8
|
Zietzer A, Düsing P, Reese L, Nickenig G, Jansen F. Ceramide Metabolism in Cardiovascular Disease: A Network With High Therapeutic Potential. Arterioscler Thromb Vasc Biol 2022; 42:1220-1228. [PMID: 36004640 DOI: 10.1161/atvbaha.122.318048] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Growing evidence suggests that ceramides play an important role in the development of atherosclerotic and valvular heart disease. Ceramides are biologically active sphingolipids that are produced by a complex network of enzymes. Lowering cellular and tissue levels of ceramide by inhibiting the ceramide-producing enzymes counteracts atherosclerotic and valvular heart disease development in animal models. In vascular tissues, ceramides are produced in response to hyperglycemia and TNF (tumor necrosis factor)-α signaling and are involved in NO-signaling and inflammation. In humans, elevated blood ceramide levels are associated with cardiovascular events. Furthermore, important cardiovascular risk factors, such as obesity and diabetes, have been linked to ceramide accumulation. This review summarizes the basic mechanisms of how ceramides drive cardiovascular disease locally and links these findings to the intriguing results of human studies on ceramides as biomarkers for cardiovascular events. Moreover, we discuss the current state of interventions to therapeutically influence vascular ceramide metabolism, both locally and systemically.
Collapse
Affiliation(s)
- Andreas Zietzer
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Germany
| | - Philip Düsing
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Germany
| | - Laurine Reese
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Germany
| | - Felix Jansen
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Germany
| |
Collapse
|
9
|
Zhao Y, Jia X, Yang X, Bai X, Lu Y, Zhu L, Cheng W, Shu M, Zhu Y, Du X, Wang L, Shu Y, Song Y, Jin S. Deacetylation of Caveolin-1 by Sirt6 induces autophagy and retards high glucose-stimulated LDL transcytosis and atherosclerosis formation. Metabolism 2022; 131:155162. [PMID: 35167876 DOI: 10.1016/j.metabol.2022.155162] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/20/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Atherosclerosis (AS) is the basis of diabetic macrovascular complications. The plasma low-density lipoprotein (LDL) particles transcytosis across endothelial cells (ECs) and deposition under the endothelium is the initiation step of AS. We previously reported that high glucose inhibits the autophagic degradation of Caveolin-1 and promote LDL transcytosis across ECs, which in turn accelerates atherosclerotic progression. Since Sirt6 is a chromatin-associated protein with deacetylation activity, whether it can regulate Caveolin-1 acetylation and regulating the autophagic degradation of Caveolin-1 remains elusive. METHODS Autophagy and histone acetylation were assessed in the umbilical cords of patients with gestational diabetes mellitus (GDM) by immunohistochemistry. An in vitro model of LDL transcytosis was established, and the role of Sirt6 in LDL transcytosis across endothelial cells was clarified. The effect of Sirt6 on the autophagic degradation of Caveolin-1 under hyperglycemic conditions was explored in a streptozotocin (STZ)-induced diabetic AS model established using the ApoE-/- mice. RESULTS Caveolin-1 and acetylated histone H3 levels were significantly increased, while LC3B and Sirt6 were downregulated in the monolayer of the vascular wall from GDM and type 2 diabetes mellitus (T2DM) patients. Immunoprecipitation assays showed that Sirt6 interacts with Caveolin-1 and specifically mediated its acetylation levels. Immuno-electron microscopy (EM) further indicated that Sirt6 overexpression triggered the autophagic lysosomal degradation of Caveolin-1. ECs-specific overexpression of Sirt6 by adeno-associated viral vector serotype 9 (AAV9) induced autophagy, reduced Caveolin-1 expression, and ameliorated atherosclerotic plaque formation in STZ-induced diabetic ApoE-/- mice. CONCLUSION Sirt6-mediated acetylation of Caveolin-1 activates its autophagic degradation and inhibits high glucose-stimulated LDL transcytosis. Thus, the Sirt6/Caveolin-1 autophagic pathway plays a crucial role in diabetic AS, and the overexpression or activation of Sirt6 is a novel therapeutic strategy.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Xiong Jia
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Xiaoyan Yang
- Department of Pharmacology, the Key Laboratory of Drug Target Researches and Pharmacodynamics Evaluation of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiangli Bai
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Yajing Lu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Lin Zhu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Wenzhuo Cheng
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Meng Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Yan Zhu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Xiaolong Du
- Department of Thyroid Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Li Wang
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Yi Song
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China.
| |
Collapse
|
10
|
Wang P, Zeng G, Yan Y, Zhang SY, Dong Y, Zhang Y, Zhang X, Liu H, Zhang Z, Jiang C, Pang Y. Disruption of adipocyte HIF-1α improves atherosclerosis through the inhibition of ceramide generation. Acta Pharm Sin B 2022; 12:1899-1912. [PMID: 35847503 PMCID: PMC9279628 DOI: 10.1016/j.apsb.2021.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/05/2021] [Accepted: 09/14/2021] [Indexed: 11/27/2022] Open
Abstract
Atherosclerosis is a chronic multifactorial cardiovascular disease. Western diets have been reported to affect atherosclerosis through regulating adipose function. In high cholesterol diet-fed ApoE–/– mice, adipocyte HIF-1α deficiency or direct inhibition of HIF-1α by the selective pharmacological HIF-1α inhibitor PX-478 alleviates high cholesterol diet-induced atherosclerosis by reducing adipose ceramide generation, which lowers cholesterol levels and reduces inflammatory responses, resulting in improved dyslipidemia and atherogenesis. Smpd3, the gene encoding neutral sphingomyelinase, is identified as a new target gene directly regulated by HIF-1α that is involved in ceramide generation. Injection of lentivirus-SMPD3 in epididymal adipose tissue reverses the decrease in ceramides in adipocytes and eliminates the improvements on atherosclerosis in the adipocyte HIF-1α-deficient mice. Therefore, HIF-1α inhibition may constitute a novel approach to slow atherosclerotic progression.
Collapse
|
11
|
Sphingomyelin Synthase Family and Phospholipase Cs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:77-86. [DOI: 10.1007/978-981-19-0394-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Sphingolipids and Cholesterol. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:1-14. [DOI: 10.1007/978-981-19-0394-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Li Z, Chiang YP, He M, Worgall TS, Zhou H, Jiang XC. Liver sphingomyelin synthase 1 deficiency causes steatosis, steatohepatitis, fibrosis, and tumorigenesis: An effect of glucosylceramide accumulation. iScience 2021; 24:103449. [PMID: 34927020 PMCID: PMC8649732 DOI: 10.1016/j.isci.2021.103449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/21/2021] [Accepted: 11/11/2021] [Indexed: 11/30/2022] Open
Abstract
Glucosylceramide (GluCer) was accumulated in sphingomyelin synthase 1 (SMS1) but not SMS2 deficient mouse tissues. In current study, we studied GluCer accumulation-mediated metabolic consequences. Livers from liver-specific Sms1/global Sms2 double-knockout (dKO) exhibited severe steatosis under a high-fat diet. Moreover, chow diet-fed ≥6-month-old dKO mice had liver impairment, inflammation, and fibrosis, compared with wild type and Sms2 KO mice. RNA sequencing showed 3- to 12-fold increases in various genes which are involved in lipogenesis, inflammation, and fibrosis. Further, we found that direct GluCer treatment (in vitro and in vivo) promoted hepatocyte to secrete more activated TGFβ1, which stimulated more collagen 1α1 production in hepatic stellate cells. Additionally, GluCer promoted more β-catenin translocation into the nucleus, thus promoting tumorigenesis. Importantly, human NASH patients had higher liver GluCer synthase and higher plasma GluCer. These findings implicated that GluCer accumulation is one of triggers promoting the development of NAFLD into NASH, then, fibrosis, and tumorigenesis.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Yeun-po Chiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Mulin He
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | | | | | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York, USA
| |
Collapse
|
14
|
Choi RH, Tatum SM, Symons JD, Summers SA, Holland WL. Ceramides and other sphingolipids as drivers of cardiovascular disease. Nat Rev Cardiol 2021; 18:701-711. [PMID: 33772258 PMCID: PMC8978615 DOI: 10.1038/s41569-021-00536-1] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 02/03/2023]
Abstract
Increases in calorie consumption and sedentary lifestyles are fuelling a global pandemic of cardiometabolic diseases, including coronary artery disease, diabetes mellitus, cardiomyopathy and heart failure. These lifestyle factors, when combined with genetic predispositions, increase the levels of circulating lipids, which can accumulate in non-adipose tissues, including blood vessel walls and the heart. The metabolism of these lipids produces bioactive intermediates that disrupt cellular function and survival. A compelling body of evidence suggests that sphingolipids, such as ceramides, account for much of the tissue damage in these cardiometabolic diseases. In humans, serum ceramide levels are proving to be accurate biomarkers of adverse cardiovascular disease outcomes. In mice and rats, pharmacological inhibition or depletion of enzymes driving de novo ceramide synthesis prevents the development of diabetes, atherosclerosis, hypertension and heart failure. In cultured cells and isolated tissues, ceramides perturb mitochondrial function, block fuel usage, disrupt vasodilatation and promote apoptosis. In this Review, we discuss the body of literature suggesting that ceramides are drivers - and not merely passengers - on the road to cardiovascular disease. Moreover, we explore the feasibility of therapeutic strategies to lower ceramide levels to improve cardiovascular health.
Collapse
Affiliation(s)
- Ran Hee Choi
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
15
|
Taniguchi M, Okazaki T. Role of ceramide/sphingomyelin (SM) balance regulated through "SM cycle" in cancer. Cell Signal 2021; 87:110119. [PMID: 34418535 DOI: 10.1016/j.cellsig.2021.110119] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
Sphingomyelin synthase (SMS), which comprises of two isozymes, SMS1 and SMS2, is the only enzyme that generates sphingomyelin (SM) by transferring phosphocholine of phosphatidylcholine to ceramide in mammals. Conversely, ceramide is generated from SM hydrolysis via sphingomyelinases (SMases), ceramide de novo synthesis, and the salvage pathway. The biosynthetic pathway for SM and ceramide content by SMS and SMase, respectively, is called "SM cycle." SM forms a SM-rich microdomain on the cell membrane to regulate signal transduction, such as proliferation/survival, migration, and inflammation. On the other hand, ceramide acts as a lipid mediator by forming a ceramide-rich platform on the membrane, and ceramide exhibits physiological actions such as cell death, cell cycle arrest, and autophagy induction. Therefore, the regulation of ceramide/SM balance by SMS and SMase is responsible for diverse cell functions not only in physiological cells but also in cancer cells. This review outlines the implications of ceramide/SM balance through "SM cycle" in cancer progression and prevention. In addition, the possible involvement of "SM cycle" is introduced in anti-cancer tumor immunity, which has become a hot topic to innovate a more effective and safer way to conquer cancer in recent years.
Collapse
Affiliation(s)
- Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0293, Japan
| | - Toshiro Okazaki
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, 1-308 Suematsu, Nonoichi-shi, Ishikawa 921-8836, Japan; Faculty of Advanced Life Science, Graduate School of Life Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo 060-0810, Japan.
| |
Collapse
|
16
|
Zhang K, Zheng J, Chen Y, Dong J, Li Z, Chiang YP, He M, Huang Q, Tang H, Jiang XC. Inducible phospholipid transfer protein deficiency ameliorates atherosclerosis. Atherosclerosis 2021; 324:9-17. [PMID: 33798923 DOI: 10.1016/j.atherosclerosis.2021.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis progression and regression studies are related to its prevention and treatment. Although we have gained extensive knowledge on germline phospholipid transfer protein (PLTP) deficiency, the effect of inducible PLTP deficiency in atherosclerosis remains unexplored. METHODS We generated inducible PLTP (iPLTP)-knockout (KO) mice and measured their plasma lipid levels after feeding a normal chow or a Western-type diet. Adenovirus associated virus-proprotein convertase subtilisin/kexin type 9 (AAV-PCSK9) was used to induce hypercholesterolemia in the mice. Collars were placed around the common carotid arteries, and atherosclerosis progression and regression in the carotid arteries and aortic roots were evaluated. RESULTS On a normal chow diet, iPLTP-KO mice exhibited decreased cholesterol, phospholipid, apoA-I, and apoB levels compared with control mice. Furthermore, the overall amount of high-density lipoprotein (HDL) particles was reduced in these mice, but this effect was more profound for larger HDL particles. On a Western-type diet, iPLTP-KO mice again exhibited reduced levels of all tested lipids, even though the basal lipid levels were increased. Additionally, these mice displayed significantly reduced atherosclerotic plaque sizes with increased plaque stability. Importantly, inducible PLTP deficiency significantly ameliorated atherosclerosis by reducing the size of established plaques and the number of macrophages in the plaques without causing lipid accumulation in the liver. CONCLUSIONS Induced PLTP deficiency in adult mice reduces plasma total cholesterol and triglycerides, prevents atherosclerosis progression, and promotes atherosclerosis regression. Thus, PLTP inhibition is a promising therapeutic approach for atherosclerosis.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiao Zheng
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Beijing University of Chinese Medicine, Beijing, China
| | | | | | - Zhiqiang Li
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA
| | - Yeun-Po Chiang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Mulin He
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | | | | | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA.
| |
Collapse
|
17
|
Ottosson F, Emami Khoonsari P, Gerl MJ, Simons K, Melander O, Fernandez C. A plasma lipid signature predicts incident coronary artery disease. Int J Cardiol 2021; 331:249-254. [PMID: 33545264 DOI: 10.1016/j.ijcard.2021.01.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Dyslipidemia is a hallmark of cardiovascular disease but is characterized by crude measurements of triglycerides, HDL- and LDL cholesterol. Lipidomics enables more detailed measurements of plasma lipids, which may help improve risk stratification and understand the pathophysiology of cardiovascular disease. METHODS Lipidomics was used to measure 184 lipids in plasma samples from the Malmö Diet and Cancer - Cardiovascular Cohort (N = 3865), taken at baseline examination. During an average follow-up time of 20.3 years, 536 participants developed coronary artery disease (CAD). Least absolute shrinkage and selection operator (LASSO) were applied to Cox proportional hazards models in order to identify plasma lipids that predict CAD. RESULTS Eight plasma lipids improved prediction of future CAD on top of traditional cardiovascular risk factors. Principal component analysis of CAD-associated lipids revealed one principal component (PC2) that was associated with risk of future CAD (HR per SD increment =1.46, C·I = 1.35-1.48, P < 0.001). The risk increase for being in the highest quartile of PC2 (HR = 2.33, P < 0.001) was higher than being in the top quartile of systolic blood pressure. Addition of PC2 to traditional risk factors achieved an improvement (2%) in the area under the ROC-curve for CAD events occurring within 10 (P = 0.03), 15 (P = 0.003) and 20 (P = 0.001) years of follow-up respectively. CONCLUSIONS A lipid pattern improve CAD prediction above traditional risk factors, highlighting that conventional lipid-measures insufficiently describe dyslipidemia that is present years before CAD. Identifying this hidden dyslipidemia may help motivate lifestyle and pharmacological interventions early enough to reach a substantial reduction in absolute risk.
Collapse
Affiliation(s)
- Filip Ottosson
- Department of Clinical Sciences, Lund University, Malmö, Sweden.
| | - Payam Emami Khoonsari
- Department of Clinical Sciences, Lund University, Malmö, Sweden; Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Box 1031, SE-17121 Solna, Sweden
| | - Mathias J Gerl
- Lipotype GmbH, Dresden, Germany; Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | |
Collapse
|
18
|
Li Z, Chiang YP, He M, Zhang K, Zheng J, Wu W, Cai J, Chen Y, Chen G, Chen Y, Dong J, Worgall TS, Jiang XC. Effect of liver total sphingomyelin synthase deficiency on plasma lipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158898. [PMID: 33545384 DOI: 10.1016/j.bbalip.2021.158898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 11/26/2022]
Abstract
Sphingomyelin (SM) is one major phospholipids on lipoproteins. It is enriched on apolipoprotein B-containing particles, including very low-density lipoprotein (VLDL) and its catabolites, low-density lipoprotein (LDL). SM is synthesized by sphingomyelin synthase 1 and 2 (SMS1 and SMS2) which utilizes ceramide and phosphatidylcholine, as two substrates, to produce SM and diacylglyceride. SMS1 and SMS2 activities are co-expressed in all tested tissues, including the liver where VLDL is produced. Thus, neither Sms1 gene knockout (KO) nor Sms2 KO approach is sufficient to evaluate the effect of SMS on VLDL metabolism. We prepared liver-specific Sms1 KO/global Sms2 KO mice to evaluate the effect of hepatocyte SM biosynthesis in lipoprotein metabolism. We found that hepatocyte total SMS depletion significantly reduces cellular sphingomyelin levels. Also, we found that the deficiency induces cellular glycosphingolipid levels which is specifically related with SMS1 but not SMS2 deficiency. To our surprise, hepatocyte total SMS deficiency has marginal effect on hepatocyte ceramide, diacylglyceride, and phosphatidylcholine levels. Importantly, total SMS deficiency decreases plasma triglyceride but not apoB levels and reduces larger VLDL concentration. The reduction of triglyceride levels also was observed when the animals were on a high fat diet. Our results show that hepatocyte total SMS blocking can reduce VLDL-triglyceride production and plasma triglyceride levels. This phenomenon could be related with a reduction of atherogenicity.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Yeun-Po Chiang
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Mulin He
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Ke Zhang
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Jiao Zheng
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Weihua Wu
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Jiajia Cai
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Yong Chen
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Guangzhi Chen
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | | | | | - Tilla S Worgall
- Department of Medicine, Columbia University, United States of America
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, United States of America.
| |
Collapse
|
19
|
Translational insight into prothrombotic state and hypercoagulation in nonalcoholic fatty liver disease. Thromb Res 2020; 198:139-150. [PMID: 33340925 DOI: 10.1016/j.thromres.2020.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/17/2020] [Accepted: 12/07/2020] [Indexed: 02/08/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an emerging and threatening pathological condition, ranging from fatty liver (FL) to chronic steatohepatitis (NASH), liver cirrhosis, and eventually to hepatocellular carcinoma (HCC). Recent findings suggest that patients with NAFLD have a higher risk of cardiovascular events and thromboembolism and that this risk is independent of metabolic diseases that are frequently associated with NAFLD, such as diabetes, hyperlipidaemia, and obesity. The vascular involvement of NAFLD might be considered its systemic burden, conditioning higher mortality in patients affected by the disease. These clinical findings suggested the existence of a prothrombotic state in NAFLD, which is partially unexplored and whose underlying mechanisms are to date not completely understood. Here, we review the mechanisms involved in the pathogenesis of the prothrombotic state in NAFLD across the progression from the healthy liver through the different stages of the disease. We focused on the possible role of several metabolic features of NAFLD possibly leading to hypercoagulation other than endothelial and platelet activation, such as insulin-resistance, nitric oxide production regulation, and gut microbiota homeostasis. Also, we analysed the involvement of plasminogen activator inhibitor-1 (PAI-1) and thromboinflammation taking place in NAFLD. Finally, we described factors striking a prothrombotic imbalance in NASH cirrhosis, with a particular focus on the pathogenesis of portal vein thrombosis.
Collapse
|
20
|
Huang J, Huth C, Covic M, Troll M, Adam J, Zukunft S, Prehn C, Wang L, Nano J, Scheerer MF, Neschen S, Kastenmüller G, Suhre K, Laxy M, Schliess F, Gieger C, Adamski J, Hrabe de Angelis M, Peters A, Wang-Sattler R. Machine Learning Approaches Reveal Metabolic Signatures of Incident Chronic Kidney Disease in Individuals With Prediabetes and Type 2 Diabetes. Diabetes 2020; 69:2756-2765. [PMID: 33024004 DOI: 10.2337/db20-0586] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/29/2020] [Indexed: 11/13/2022]
Abstract
Early and precise identification of individuals with prediabetes and type 2 diabetes (T2D) at risk for progressing to chronic kidney disease (CKD) is essential to prevent complications of diabetes. Here, we identify and evaluate prospective metabolite biomarkers and the best set of predictors of CKD in the longitudinal, population-based Cooperative Health Research in the Region of Augsburg (KORA) cohort by targeted metabolomics and machine learning approaches. Out of 125 targeted metabolites, sphingomyelin C18:1 and phosphatidylcholine diacyl C38:0 were identified as candidate metabolite biomarkers of incident CKD specifically in hyperglycemic individuals followed during 6.5 years. Sets of predictors for incident CKD developed from 125 metabolites and 14 clinical variables showed highly stable performances in all three machine learning approaches and outperformed the currently established clinical algorithm for CKD. The two metabolites in combination with five clinical variables were identified as the best set of predictors, and their predictive performance yielded a mean area value under the receiver operating characteristic curve of 0.857. The inclusion of metabolite variables in the clinical prediction of future CKD may thus improve the risk prediction in people with prediabetes and T2D. The metabolite link with hyperglycemia-related early kidney dysfunction warrants further investigation.
Collapse
Affiliation(s)
- Jialing Huang
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Cornelia Huth
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Marcela Covic
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Martina Troll
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Jonathan Adam
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Sven Zukunft
- Research Unit of Molecular Endocrinology and Metabolism, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Cornelia Prehn
- Research Unit of Molecular Endocrinology and Metabolism, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Li Wang
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Scientific Research and Shandong University Postdoctoral Work Station, Liaocheng People's Hospital, Shandong, P. R. China
| | - Jana Nano
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Markus F Scheerer
- Institute of Experimental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Susanne Neschen
- Institute of Experimental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine - Qatar, Doha, Qatar
| | - Michael Laxy
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Jerzy Adamski
- Research Unit of Molecular Endocrinology and Metabolism, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising, Germany
| | - Martin Hrabe de Angelis
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Rui Wang-Sattler
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
21
|
Taniguchi M, Okazaki T. Ceramide/Sphingomyelin Rheostat Regulated by Sphingomyelin Synthases and Chronic Diseases in Murine Models. J Lipid Atheroscler 2020; 9:380-405. [PMID: 33024732 PMCID: PMC7521967 DOI: 10.12997/jla.2020.9.3.380] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 12/16/2022] Open
Abstract
Ceramide and sphingomyelin (SM) are major components of the double membrane-bound sphingolipids. Ceramide is an essential bioactive lipid involved in numerous cell processes including apoptosis, necrosis, and autophagy-dependent cell death. Inversely, SM regulates opposite cellular processes such as proliferation and migration by changing receptor-mediated signal transduction in the lipid microdomain. SM is generated through a transfer of phosphocholine from phosphatidylcholine to ceramide by SM synthases (SMSs). Research during the past several decades has revealed that the ceramide/SM balance in cellular membranes regulated by SMSs is important to decide the cell fate, survival, and proliferation. In addition, recent experimental studies utilizing SMS knockout mice and murine disease models provide evidence that SMS-regulated ceramide/SM balance is involved in human diseases. Here, we review the basic structural and functional characteristics of SMSs and focus on their cellular functions through the regulation of ceramide/SM balance in membrane microdomains. In addition, we present the pathological or physiological implications of SMSs by analyzing their role in SMS-knockout mice and human disease models. This review finally presents evidence indicating that the regulation of ceramide/SM balance through SMS could be a therapeutic target for human disorders.
Collapse
Affiliation(s)
- Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku, Japan
| | - Toshiro Okazaki
- Research Institute for Bioresources and Biotechnology, Kanazawa Prefectural University, Nonoichi, Japan
| |
Collapse
|
22
|
Wang X, Wang Y, Xu J, Xue C. Sphingolipids in food and their critical roles in human health. Crit Rev Food Sci Nutr 2020; 61:462-491. [PMID: 32208869 DOI: 10.1080/10408398.2020.1736510] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sphingolipids (SLs) are ubiquitous structural components of cell membranes and are essential for cell functions under physiological conditions or during disease progression. Abundant evidence supports that SLs and their metabolites, including ceramide (Cer), ceramide-1-phosphate (C1P), sphingosine (So), sphingosine-1-phosphate (S1P), are signaling molecules that regulate a diverse range of cellular processes and human health. However, there are limited reviews on the emerging roles of exogenous dietary SLs in human health. In this review, we discuss the ubiquitous presence of dietary SLs, highlighting their structures and contents in foodstuffs, particularly in sea foods. The digestion and metabolism of dietary SLs is also discussed. Focus is given to the roles of SLs in both the etiology and prevention of diseases, including bacterial infection, cancers, neurogenesis and neurodegenerative diseases, skin integrity, and metabolic syndrome (MetS). We propose that dietary SLs represent a "functional" constituent as emerging strategies for improving human health. Gaps in research that could be of future interest are also discussed.
Collapse
Affiliation(s)
- Xiaoxu Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Yuming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.,Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.,Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| |
Collapse
|
23
|
Taniguchi M, Ueda Y, Matsushita M, Nagaya S, Hashizume C, Arai K, Kabayama K, Fukase K, Watanabe K, Wardhani LO, Hayashi K, Okazaki T. Deficiency of sphingomyelin synthase 2 prolongs survival by the inhibition of lymphoma infiltration through ICAM-1 reduction. FASEB J 2020; 34:3838-3854. [PMID: 31970839 DOI: 10.1096/fj.201901783rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/10/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022]
Abstract
The tumor microenvironment (TME) formation involving host cells and cancer cells through cell adhesion molecules (CAMs) is essential for the multiple steps of cancer metastasis and growth. Sphingomyelin synthase 2 (SMS2) is involved in inflammatory diseases such as obesity and diabetes mellitus by regulation of the SM/ceramide balance. However, the involvement of SMS2 in TME formation and metastasis is largely unknown. Here, we report that SMS2-deficient (SMS2-KO) mice show suppressed the EL4 cell infiltration to liver and prolonged survival time. ICAM-1 was identified as a candidate for the inhibition of TME formation in immortalized mouse embryonic fibroblasts (tMEFs) from mRNA array analysis for CAMs. Reduced SM/ceramide balance in SMS2-KO tMEFs suppressed the attachment of EL4 cells through transcriptional reduction of ICAM-1 by the inhibition of NF-κB activation. TNF-α-induced NF-κB activation and subsequent induction of ICAM-1 were suppressed in SMS2-KO tMEFs but restored by SMS2 re-introduction. In the EL4 cell infiltration mouse model, EL4 injection increased ICAM-1 expression in WT liver but not in SMS2-KO mouse liver. Therefore, inhibition of SMS2 may be a therapeutic target to suppress the infiltration of malignant lymphoma.
Collapse
Affiliation(s)
- Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku, Japan
| | - Yoshibumi Ueda
- Faculty of Medicine, Department of Hematology and Immunology, Kanazawa Medical University, Kahoku, Japan.,Graduate School of Arts and Sciences, University of Tokyo, Tokyo, Japan
| | - Michiko Matsushita
- Department of Pathobiological Science and Technology, School of Health Science, University of Tottori, Tottori, Japan
| | - Shingo Nagaya
- Faculty of Medicine, Department of Hematology and Immunology, Kanazawa Medical University, Kahoku, Japan
| | - Chieko Hashizume
- Faculty of Medicine, Department of Hematology and Immunology, Kanazawa Medical University, Kahoku, Japan
| | - Kenta Arai
- Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan.,Project Research Center, Graduate School of Science, Osaka University, Osaka, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan.,Project Research Center, Graduate School of Science, Osaka University, Osaka, Japan
| | - Ken Watanabe
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Lusi Oka Wardhani
- Division of Molecular Pathology, Faculty of Medicine, Department of Microbiology and Pathology, Tottori University, Tottori, Japan
| | - Kazuhiko Hayashi
- Division of Molecular Pathology, Faculty of Medicine, Department of Microbiology and Pathology, Tottori University, Tottori, Japan
| | - Toshiro Okazaki
- Faculty of Medicine, Department of Hematology and Immunology, Kanazawa Medical University, Kahoku, Japan.,Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Kahoku, Japan
| |
Collapse
|
24
|
Koolath S, Murai Y, Suga Y, Monde K. Chiral combinatorial preparation and biological evaluation of unique ceramides for inhibition of sphingomyelin synthase. Chirality 2020; 32:308-313. [DOI: 10.1002/chir.23179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 12/23/2019] [Accepted: 12/26/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Sajeer Koolath
- Graduate School of Life ScienceHokkaido University Sapporo Japan
| | - Yuta Murai
- Graduate School of Life ScienceHokkaido University Sapporo Japan
- Faculty of Advanced Life ScienceHokkaido University Sapporo Japan
| | - Yoshiko Suga
- Faculty of Advanced Life ScienceHokkaido University Sapporo Japan
| | - Kenji Monde
- Graduate School of Life ScienceHokkaido University Sapporo Japan
- Faculty of Advanced Life ScienceHokkaido University Sapporo Japan
| |
Collapse
|
25
|
Othman MA, Yuyama K, Murai Y, Igarashi Y, Mikami D, Sivasothy Y, Awang K, Monde K. Malabaricone C as Natural Sphingomyelin Synthase Inhibitor against Diet-Induced Obesity and Its Lipid Metabolism in Mice. ACS Med Chem Lett 2019; 10:1154-1158. [PMID: 31413799 DOI: 10.1021/acsmedchemlett.9b00171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/03/2019] [Indexed: 01/16/2023] Open
Abstract
The interaction between natural occurring inhibitors and targeted membrane proteins could be an alternative medicinal strategy for the treatment of metabolic syndrome, notably, obesity. In this study, we identified malabaricones A-C and E (1-4) isolated from the fruits of Myristica cinnamomea King as natural inhibitors for sphingomyelin synthase (SMS), a membrane protein responsible for sphingolipid biosynthesis. Having the most promising inhibition, oral administration of compound 3 exhibited multiple efficacies in reducing weight gain, improving glucose tolerance, and reducing hepatic steatosis in high fat diet-induced obesity mice models. Liver lipid analysis revealed a crucial link between the SMS activities of compound 3 and its lipid metabolism in vitro and in vivo. The nontoxic nature of compound 3 makes it a suitable candidate in search of drugs which can be employed in the treatment and prevention of obesity.
Collapse
Affiliation(s)
- Muhamad Aqmal Othman
- Graduate School of Life Science, Hokkaido University, Kita 21 Nishi 11, Sapporo 001-0021, Japan
| | - Kohei Yuyama
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Kita 21 Nishi 11, Sapporo, 001-0021, Japan
| | - Yuta Murai
- Faculty of Advanced Life Science, Hokkaido University, Kita 21 Nishi 11, Sapporo 001-0021, Japan
| | - Yasuyuki Igarashi
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Kita 21 Nishi 11, Sapporo, 001-0021, Japan
| | - Daisuke Mikami
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Kita 21 Nishi 11, Sapporo, 001-0021, Japan
| | - Yasodha Sivasothy
- Research Centre for Crystalline Materials, Faculty of Science and Technology, Sunway University, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| | - Khalijah Awang
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kenji Monde
- Faculty of Advanced Life Science, Hokkaido University, Kita 21 Nishi 11, Sapporo 001-0021, Japan
| |
Collapse
|
26
|
Shi XX, Zhang H, Chen M, Zhang YD, Zhu MF, Zhang MJ, Li FQ, Wratten S, Zhou WW, Mao C, Zhu ZR. Two sphingomyelin synthase homologues regulate body weight and sphingomyelin synthesis in female brown planthopper, N. lugens (Stål). INSECT MOLECULAR BIOLOGY 2019; 28:253-263. [PMID: 30375099 DOI: 10.1111/imb.12549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Although sphingomyelins known to be are lipid constituents of the plasma membrane in vertebrates, much remains obscure about the metabolism of sphingomyelins in insects. With ultra performance liquid chromatography-time-of-flight-tandem mass spectrometry analysis, we revealed for the first time that sphingomyelins are abundant in Nilaparvata lugens (Stål), the brown planthopper (BPH), and their biosynthesis is carried out by sphingomyelin synthase-like protein 2 (SMSL2), which is homologous to sphingomyelin synthase-related protein (SMSr). Unlike other insect species, high concentrations of sphingomyelins rather than ceramide phosphoethanolamines exist in the BPH. Two putative genes, which are homologous to SMSr, are named Nilaparvata lugens SMS-like 1 (NlSMSL1) and 2 (NlSMSL2). Knockdowns of both NlSMSL2 and NlSMSL1 were conducted but only the first decreased concentrations of sphingomyelins in the BPH, indicating that NlSMSL2 plays a role in the biosynthesis of sphingomyelins. Real-time quantitative PCR analysis revealed both NlSMSL1 and NlSMSL2 are highly expressed in BPH adults, with NlSMSL1 specifically highly expressed in reproductive organs (ovaries and testes) whereas NlSMSL2 was highly expressed in the malpighian tubules. The knockdown of NlSMSL1 or NlSMSL2 increased BPH female body weight but not that of males, suggesting sex-specific roles for SMSLs in influencing BPH body weight. The results suggest that NlSMSL2 catalyses the synthesis of sphingomyelins and maintains female BPH body weight through alteration of sphingolipid content.
Collapse
Affiliation(s)
- X-X Shi
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
- Institute of Insect Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - H Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M Chen
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - Y-D Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M-F Zhu
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M-J Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - F-Q Li
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - S Wratten
- Bio-Protection Research Centre, Lincoln University, Lincoln, New Zealand
| | - W-W Zhou
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - C Mao
- Department of Medicine and Stony Brook Cancer Center, The State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Z-R Zhu
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Potential therapeutic targets for atherosclerosis in sphingolipid metabolism. Clin Sci (Lond) 2019; 133:763-776. [PMID: 30890654 PMCID: PMC6422862 DOI: 10.1042/cs20180911] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022]
Abstract
Sphingolipids, such as sphingomyelins, ceramides, glycosphingolipids, and sphingosine-1-phosphates (S1P) are a large group of structurally and functionally diverse molecules. Some specific species are found associated with atherogenesis and provide novel therapeutic targets. Herein, we briefly review how sphingolipids are implicated in the progression of atherosclerosis and related diseases, and then we discuss the potential therapy options by targetting several key enzymes in sphingolipid metabolism.
Collapse
|
28
|
Rauschert S, Gázquez A, Uhl O, Kirchberg FF, Demmelmair H, Ruíz-Palacios M, Prieto-Sánchez MT, Blanco-Carnero JE, Nieto A, Larqué E, Koletzko B. Phospholipids in lipoproteins: compositional differences across VLDL, LDL, and HDL in pregnant women. Lipids Health Dis 2019; 18:20. [PMID: 30670033 PMCID: PMC6343318 DOI: 10.1186/s12944-019-0957-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 01/01/2019] [Indexed: 12/11/2022] Open
Abstract
Objective The aim of this study was to analyse the differences in the phospholipid composition of very low density (VLDL), low density (LDL) and high density lipoprotein (HDL) monolayers in pregnant lean and obese women. Methods LDL, HDL, and VLDL were isolated from plasma samples of 10 lean and 10 obese pregnant women, and their species composition of phosphatidylcholines (PC) and sphingomyelins (SM) was analysed by liquid-chromatography tandem mass-spectrometry. Wilcoxon-Mann-Whitney U test and principal component analysis (PCA) were used to investigate if metabolite profiles differed between the lean/obese group and between lipoprotein species. Results No significant differences have been found in the metabolite levels between obese and non-obese pregnant women. The PCA components 1 and 2 separated between LDL, HDL, and VLDL but not between normal weight and obese women. Twelve SM and one PCae were more abundant in LDL than in VLDL. In contrast, four acyl-alkyl-PC and two diacyl-PC were significantly higher in HDL compared to LDL. VLDL and HDL differed in three SM, seven acyl-alkyl-PC and one diacyl-PC (higher values in HDL) and 13 SM (higher in VLDL). We also found associations of some phospholipid species with HDL and LDL cholesterol. Conclusion In pregnant women phospholipid composition differs significantly in HDL, LDL and VLDL, similar to previous findings in men and non-pregnant women. Obese and lean pregnant women showed no significant differences in their lipoprotein associated metabolite profile. Electronic supplementary material The online version of this article (10.1186/s12944-019-0957-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sebastian Rauschert
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany
| | - Antonio Gázquez
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany.,Department of Physiology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - Olaf Uhl
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany
| | - Franca F Kirchberg
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany
| | - Hans Demmelmair
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany
| | - María Ruíz-Palacios
- Department of Physiology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - María T Prieto-Sánchez
- Obstetrics and Gynecology Service, Virgen de la Arrixaca Clinical Hospital, University of Murcia, Murcia, Murcia, Spain
| | - José E Blanco-Carnero
- Obstetrics and Gynecology Service, Virgen de la Arrixaca Clinical Hospital, University of Murcia, Murcia, Murcia, Spain
| | - Anibal Nieto
- Obstetrics and Gynecology Service, Virgen de la Arrixaca Clinical Hospital, University of Murcia, Murcia, Murcia, Spain
| | - Elvira Larqué
- Department of Physiology, Faculty of Biology, University of Murcia, Murcia, Spain
| | - Berthold Koletzko
- LMU - Ludwig-Maximilians-Universität Munich, Div. Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337, Munich, Germany.
| |
Collapse
|
29
|
Discovery, synthesis and anti-atherosclerotic activities of a novel selective sphingomyelin synthase 2 inhibitor. Eur J Med Chem 2018; 163:864-882. [PMID: 30580239 DOI: 10.1016/j.ejmech.2018.12.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 02/03/2023]
Abstract
The sphingomyelin synthase 2 (SMS2) is a potential target for pharmacological intervention in atherosclerosis. However, so far, few selective SMS2 inhibitors and their pharmacological activities were reported. In this study, a class of 2-benzyloxybenzamides were discovered as novel SMS2 inhibitors through scaffold hopping and structural optimization. Among them, Ly93 as one of the most potent inhibitors exhibited IC50 values of 91 nM and 133.9 μM against purified SMS2 and SMS1 respectively. The selectivity ratio of Ly93 was more than 1400-fold for purified SMS2 over SMS1. The in vitro studies indicated that Ly93 not only dose-dependently diminished apoB secretion from Huh7 cells, but also significantly reduced the SMS activity and increased cholesterol efflux from macrophages. Meanwhile, Ly93 inhibited the secretion of LPS-mediated pro-inflammatory cytokine and chemokine in macrophages. The pharmacokinetic profiles of Ly93 performed on C57BL/6J mice demonstrated that Ly93 was orally efficacious. As a potent selective SMS2 inhibitor, Ly93 significantly decreased the plasma SM levels of C57BL/6J mice. Furthermore, Ly93 was capable of dose-dependently attenuating the atherosclerotic lesions in the root and the entire aorta as well as macrophage content in lesions, in apolipoprotein E gene knockout mice treated with Ly93. In conclusion, we discovered a novel selective SMS2 inhibitor Ly93 and demonstrated its anti-atherosclerotic activities in vivo. The preliminary molecular mechanism-of-action studies revealed its function in lipid homeostasis and inflammation process, which indicated that the selective inhibition of SMS2 would be a promising treatment for atherosclerosis.
Collapse
|
30
|
Lou B, Liu Q, Hou J, Kabir I, Liu P, Ding T, Dong J, Mo M, Ye D, Chen Y, Bui HH, Roth K, Cao Y, Jiang XC. 2-Hydroxy-oleic acid does not activate sphingomyelin synthase activity. J Biol Chem 2018; 293:18328-18336. [PMID: 30305392 DOI: 10.1074/jbc.ra118.005904] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/02/2018] [Indexed: 11/06/2022] Open
Abstract
2-Hydroxy-oleic acid (2OHOA) is a potent anticancer drug that induces cancer cell cycle arrest and apoptosis. Previous studies have suggested that 2OHOA's anticancer effect is mediated by SMS activation in cancer cells, including A549 and U118 cells. To confirm this phenomenon, in this study, we treated both A549 and U118 cells with 2OHOA and measured SMS activity. To our surprise, we found neither 2OHOA-mediated SMS activation nor sphingomyelin accumulation in the cells. However, we noted that 2OHOA significantly reduces phosphatidylcholine in these cells. We also did not observe 2OHOA-mediated SMS activation in mouse tissue homogenates. Importantly, 2OHOA inhibited rather than activated recombinant SMS1 (rSMS1) and rSMS2 in a dose-dependent fashion. Intra-gastric treatment of C57BL/6J mice with 2OHOA for 10 days had no effects on liver and small intestine SMS activities and plasma sphingomyelin levels. The treatment inhibited lysophosphatidylcholine acyltransferase (LPCAT) activity, consistent with the aforementioned reduction in plasma phosphatidylcholine. Because total cellular phosphatidylcholine is used as a predictive biomarker for monitoring tumor responses, the previously reported 2OHOA-mediated cancer suppression could be related to this phosphatidylcholine reduction, which may influence cell membrane structure and properties. We conclude that 2OHOA is not a SMS activator and that its anticancer property may be related to an effect on phosphatidylcholine metabolism.
Collapse
Affiliation(s)
- Bin Lou
- From the School of Pharmacy, Fudan University, Shanghai 201203, China,.
| | - Qi Liu
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiahui Hou
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Inamul Kabir
- the Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203
| | - Peipei Liu
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tingbo Ding
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jibin Dong
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mingguang Mo
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Deyong Ye
- From the School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yang Chen
- the Institute of Precision Medicine, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China
| | - Hai H Bui
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, Indiana 46285, and
| | - Kenneth Roth
- Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, Indiana 46285, and
| | - Yu Cao
- the Institute of Precision Medicine, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China,.
| | - Xian-Cheng Jiang
- From the School of Pharmacy, Fudan University, Shanghai 201203, China,; the Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203,; the Molecular and Cellular Cardiology Program, Veterans Affairs New York Harbor Healthcare System, Brooklyn, New York 11209
| |
Collapse
|
31
|
Mo M, Yang J, Jiang XC, Cao Y, Fei J, Chen Y, Qi X, Chu Y, Zhou L, Ye D. Discovery of 4-Benzyloxybenzo[ d]isoxazole-3-amine Derivatives as Highly Selective and Orally Efficacious Human Sphingomyelin Synthase 2 Inhibitors that Reduce Chronic Inflammation in db/ db Mice. J Med Chem 2018; 61:8241-8254. [PMID: 30074791 DOI: 10.1021/acs.jmedchem.8b00727] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sphingomyelin synthase 2 (SMS2) is a promising therapeutic target for several chronic inflammation-associated diseases, including atherosclerosis, fatty liver, and insulin resistance. Herein, we report the identification of 4-benzyloxybenzo[ d]isoxazole-3-amine derivatives as potent and highly selective SMS2 inhibitors through a conformational restriction strategy. After systematic structural modifications, several compounds with high selectivity and good potency in vitro were selected for further evaluation. Compound 15w demonstrated good pharmacokinetics (oral bioavailability, F = 56%) in vivo and has an inhibitory potency against sphingomyelin synthase activity when Institute of Cancer Research mice are provided with an oral dose of this compound. In addition, compound 15w attenuated chronic inflammation significantly in db/ db mice after oral dosing for 6 weeks.
Collapse
Affiliation(s)
- Mingguang Mo
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Jintong Yang
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | | | - Yu Cao
- Institute of Precision Medicine , Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200125 , China
| | - Jinyu Fei
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Yang Chen
- Institute of Precision Medicine , Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200125 , China
| | - Xiangyu Qi
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Yong Chu
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Lu Zhou
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Deyong Ye
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| |
Collapse
|
32
|
Swamy MMM, Murai Y, Ohno Y, Jojima K, Kihara A, Mitsutake S, Igarashi Y, Yu J, Yao M, Suga Y, Anetai M, Monde K. Structure-inspired design of a sphingolipid mimic sphingosine-1-phosphate receptor agonist from a naturally occurring sphingomyelin synthase inhibitor. Chem Commun (Camb) 2018; 54:12758-12761. [DOI: 10.1039/c8cc05595e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A monophosphate derivative of ginkgolic acid binds to sphingosine 1-phosphate (S1P) receptors to perform similar functions to the lipid mediator S1P.
Collapse
|
33
|
Chung RWS, Wang Z, Bursill CA, Wu BJ, Barter PJ, Rye KA. Effect of long-term dietary sphingomyelin supplementation on atherosclerosis in mice. PLoS One 2017; 12:e0189523. [PMID: 29240800 PMCID: PMC5730175 DOI: 10.1371/journal.pone.0189523] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022] Open
Abstract
Sphingomyelin (SM) levels in the circulation correlate positively with atherosclerosis burden. SM is a ubiquitous component of human diets, but it is unclear if dietary SM increases circulating SM levels. Dietary choline increases atherosclerosis by raising circulating trimethylamine N-oxide (TMAO) levels in mice and humans. As SM has a choline head group, we ask in this study if dietary SM accelerates atherosclerotic lesion development by increasing circulating SM and TMAO levels. Three studies were performed: (Study 1) C57BL/6 mice were maintained on a high fat diet with or without SM supplementation for 4 weeks prior to quantification of serum TMAO and SM levels; (Study 2) atherosclerosis was studied in apoE-/- mice after 16 weeks of a high fat diet without or with SM supplementation and (Study 3) apoE-/- mice were maintained on a chow diet for 19 weeks without or with SM supplementation and antibiotic treatment prior to quantification of atherosclerotic lesions and serum TMAO and SM levels. SM consumption did not increase circulating SM levels or atherosclerosis in high fat-fed apoE-/- mice. Serum TMAO levels in C57BL/6 mice were low and had no effect atherosclerosis lesion development. Dietary SM supplementation significantly reduced atherosclerotic lesion area in the aortic arch of chow-fed apoE-/- mice. This study establishes that dietary SM does not affect circulating SM levels or increase atherosclerosis in high fat-fed apoE-/- mice, but it is anti-atherogenic in chow-fed apoE-/- mice.
Collapse
Affiliation(s)
| | - Zeneng Wang
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, United States of America
| | | | - Ben J. Wu
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Philip J. Barter
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Kerry-Anne Rye
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|
34
|
Qi XY, Cao Y, Li YL, Mo MG, Zhou L, Ye DY. Discovery of the selective sphingomyelin synthase 2 inhibitors with the novel structure of oxazolopyridine. Bioorg Med Chem Lett 2017; 27:3511-3515. [DOI: 10.1016/j.bmcl.2017.05.074] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 11/30/2022]
|
35
|
Ohnishi T, Hashizume C, Taniguchi M, Furumoto H, Han J, Gao R, Kinami S, Kosaka T, Okazaki T. Sphingomyelin synthase 2 deficiency inhibits the induction of murine colitis-associated colon cancer. FASEB J 2017; 31:3816-3830. [PMID: 28522594 DOI: 10.1096/fj.201601225rr] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/24/2017] [Indexed: 12/13/2022]
Abstract
Sphingomyelin synthase 2 (SMS2) is the synthetic enzyme of sphingomyelin (SM), which regulates membrane fluidity and microdomain structure. SMS2 plays a role in LPS-induced lung injury and inflammation; however, its role in inflammation-mediated tumorigenesis is unclear. We investigated the effect of SMS2 deficiency on dextran sodium sulfate (DSS)-induced murine colitis and found inhibition of DSS-induced inflammation in SMS2-deficient (SMS2-/-) mice. DSS treatment induced a significant increase in ceramide levels, with a decrease of SM levels in SMS2-/- colon tissue, and demonstrated attenuation of the elevation of both inflammation-related gene expression and proinflammatory cytokines and chemokines, leukocyte infiltration, and MAPK and signal transducer and activator of transcription 3 activation. After undergoing transplantation of wild-type bone marrow, SMS2-/- mice also exhibited inhibition of DSS-induced inflammation in the colon, which suggested that SMS2 deficiency in bone marrow-derived immune cells was not involved in the inhibition of colitis. Finally, in an azoxymethane/DSS-induced cancer model, SMS2 deficiency significantly decreased tumor incidence in the colon. Our results demonstrate that SMS2 deficiency inhibits DSS-induced colitis and subsequent colitis-associated colon cancer via inhibition of colon epithelial cell-mediated inflammation; therefore, inhibition of SMS2 may be a potential therapeutic target for human colitis and colorectal cancer.-Ohnishi, T., Hashizume, C., Taniguchi, M., Furumoto, H., Han, J., Gao, R., Kinami, S., Kosaka, T., Okazaki, T. Sphingomyelin synthase 2 deficiency inhibits the induction of murine colitis-associated colon cancer.
Collapse
Affiliation(s)
- Toshio Ohnishi
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Chieko Hashizume
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Hidehiro Furumoto
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Jia Han
- Division of Medical Oncology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Rongfen Gao
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Shinichi Kinami
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Takeo Kosaka
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Toshiro Okazaki
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan; .,Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
36
|
Adachi R, Ogawa K, Matsumoto SI, Satou T, Tanaka Y, Sakamoto J, Nakahata T, Okamoto R, Kamaura M, Kawamoto T. Discovery and characterization of selective human sphingomyelin synthase 2 inhibitors. Eur J Med Chem 2017; 136:283-293. [PMID: 28505533 DOI: 10.1016/j.ejmech.2017.04.067] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/10/2017] [Accepted: 04/24/2017] [Indexed: 10/19/2022]
Abstract
Sphingomyelin synthase (SMS) is a membrane enzyme that catalyzes the synthesis of sphingomyelin, is required for the maintenance of plasma membrane microdomain fluidity, and has two isoforms: SMS1 and SMS2. Although these isoforms exhibit the same SMS activity, they are different enzymes with distinguishable subcellular localizations. It was reported that SMS2 KO mice displayed lower inflammatory responses and anti-atherosclerotic effects, suggesting that inhibition of SMS2 would be a potential therapeutic approach for controlling inflammatory responses and atherosclerosis. This study aimed to discover a novel small-molecule compound that selectively inhibits SMS2 enzymatic activity. We developed a human SMS2 enzyme assay with a high-throughput mass spectrometry-based screening system. We characterized the enzymatic properties of SMS2 and established a high-throughput screening-compatible assay condition. To identify human SMS2 inhibitors, we conducted compound screening using the enzyme assay. We identified a 2-quinolone derivative as a SMS2 selective inhibitor with an IC50 of 950 nM and >100-fold selectivity for SMS2 over SMS1. The 2-quinolone exhibited efficacy in a cell-based engagement assay. We demonstrated that a more potent derivative directly bound to SMS2-expressing membrane fractions in an affinity selection mass spectrometry assay. Mutational analyses revealed that the interaction of the inhibitor with SMS2 required the presence of the amino acids S227 and H229, which are located in the catalytic domain of SMS2. In conclusion, we discovered novel SMS2-selective inhibitors. 2-Quinolone SMS2 inhibitors are considered applicable for leading optimization studies. Further investigations using these SMS2 inhibitors would provide validation tools for SMS2-relevant pathways in vitro and in vivo.
Collapse
Affiliation(s)
- Ryutaro Adachi
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Kazumasa Ogawa
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Shin-Ichi Matsumoto
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takuya Satou
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yukiya Tanaka
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Jyunichi Sakamoto
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takashi Nakahata
- CVM Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Rei Okamoto
- CVM Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masahiro Kamaura
- Medicinal Chemistry Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomohiro Kawamoto
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
37
|
Meikle PJ, Summers SA. Sphingolipids and phospholipids in insulin resistance and related metabolic disorders. Nat Rev Endocrinol 2017; 13:79-91. [PMID: 27767036 DOI: 10.1038/nrendo.2016.169] [Citation(s) in RCA: 301] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Obesity, insulin resistance, type 2 diabetes mellitus and cardiovascular disease form a metabolic disease continuum that has seen a dramatic increase in prevalence in developed and developing countries over the past two decades. Dyslipidaemia resulting from hypercaloric diets is a major contributor to the pathogenesis of metabolic disease, and lipid-lowering therapies are the main therapeutic option for this group of disorders. However, the fact that dysfunctional lipid metabolism extends far beyond cholesterol and triglycerides is becoming increasingly clear. Lipidomic studies and mouse models are helping to explain the complex interactions between diet, lipid metabolism and metabolic disease. These studies are not only improving our understanding of this complex biology, but are also identifying potential therapeutic avenues to combat this growing epidemic. This Review examines what is currently known about phospholipid and sphingolipid metabolism in the setting of obesity and how metabolic pathways are being modulated for therapeutic effect.
Collapse
Affiliation(s)
- Peter J Meikle
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004, Australia
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, 201 Presidents Circle, Salt Lake City, Utah, 84112, USA
| |
Collapse
|
38
|
Adada M, Luberto C, Canals D. Inhibitors of the sphingomyelin cycle: Sphingomyelin synthases and sphingomyelinases. Chem Phys Lipids 2016. [DOI: 10.1016/j.chemphyslip.2015.07.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
39
|
Schiffmann R. The consequences of genetic and pharmacologic reduction in sphingolipid synthesis. J Inherit Metab Dis 2015; 38:77-84. [PMID: 25164785 DOI: 10.1007/s10545-014-9758-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/28/2014] [Accepted: 07/31/2014] [Indexed: 10/24/2022]
Abstract
A new therapy based on substrate synthesis reduction in sphingolipidoses is showing promise. The consequences of decreasing sphingolipid synthesis depend on the level at which synthetic blockage occurs and on the extent of the blockage. Complete synthetic blockage may be lethal if it includes all sphingolipids, such as in a global knockout of serine palmitoyltransferase. Partial inhibition of sphingolipid synthetic pathways is usually benign and may have beneficial effects in a number of lysosomal diseases and in more common pathologies, as seen in animal models for atherosclerosis, polycystic kidney disease, diabetes, and asthma. Studies of various forms of sphingolipid synthesis reduction serve to highlight not only the cellular role of these lipids but also the potential risks and therapeutic benefits of pharmacological agents to be used in therapy for human diseases.
Collapse
Affiliation(s)
- Raphael Schiffmann
- Institute of Metabolic Disease, Baylor Research Institute, 3812 Elm Street, Dallas, TX, USA,
| |
Collapse
|
40
|
Bian F, Yang X, Zhou F, Wu PH, Xing S, Xu G, Li W, Chi J, Ouyang C, Zhang Y, Xiong B, Li Y, Zheng T, Wu D, Chen X, Jin S. C-reactive protein promotes atherosclerosis by increasing LDL transcytosis across endothelial cells. Br J Pharmacol 2014; 171:2671-84. [PMID: 24517733 DOI: 10.1111/bph.12616] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/30/2013] [Accepted: 10/01/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE The retention of plasma low-density lipoprotein (LDL) particles in subendothelial space following transcytosis across the endothelium is the initial step of atherosclerosis. Whether or not C-reactive protein (CRP) can directly affect the transcytosis of LDL is not clear. Here we have examined the effect of CRP on transcytosis of LDL across endothelial cells and have explored the underlying mechanisms. EXPERIMENTAL APPROACH Effects of CRP on transcytosis of FITC-labelled LDL were examined with human umbilical vein endothelial cells and venous rings in vitro and, in vivo, ApoE(-/-) mice. Laser scanning confocal microscopy, immunohistochemistry and Oil Red O staining were used to assay LDL. KEY RESULTS CRP increased transcytosis of LDL. An NADPH oxidase inhibitor, diphenylene iodonium, and the reducing agent, dithiothreitol partly or completely blocked CRP-stimulated increase of LDL transcytosis. The PKC inhibitor, bisindolylmaleimide I and the Src kinase inhibitor, PP2, blocked the trafficking of the molecules responsible for transcytosis. Confocal imaging analysis revealed that CRP stimulated LDL uptake by endothelial cells and vessel walls. In ApoE(-/-) mice, CRP significantly promoted early changes of atherosclerosis, which were blocked by inhibitors of transcytosis. CONCLUSIONS AND IMPLICATIONS CRP promoted atherosclerosis by directly increasing the transcytosis of LDL across endothelial cells and increasing LDL retention in vascular walls. These actions of CRP were associated with generation of reactive oxygen species, activation of PKC and Src, and translocation of caveolar or soluble forms of the N-ethylmaleimide-sensitive factor attachment protein.
Collapse
Affiliation(s)
- Fang Bian
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, The Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Go YM, Kim CW, Walker DI, Kang DW, Kumar S, Orr M, Uppal K, Quyyumi AA, Jo H, Jones DP. Disturbed flow induces systemic changes in metabolites in mouse plasma: a metabolomics study using ApoE⁻/⁻ mice with partial carotid ligation. Am J Physiol Regul Integr Comp Physiol 2014; 308:R62-72. [PMID: 25377480 DOI: 10.1152/ajpregu.00278.2014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Disturbed blood flow (d-flow) occurring in branched and curved arteries promotes endothelial dysfunction and atherosclerosis, in part, by altering gene expression and epigenomic profiles in endothelial cells. While a systemic metabolic change is known to play a role in atherosclerosis, it is unclear whether it can be regulated by local d-flow. Here, we tested this hypothesis by carrying out a metabolomics study using blood plasma samples obtained from ApoE(-/-) mice that underwent a partial carotid ligation surgery to induce d-flow. Mice receiving sham ligation were used as a control. To study early metabolic changes, samples collected from 1 wk after partial ligation when endothelial dysfunction occurs, but before atheroma develops, were analyzed by high-resolution mass spectrometry. A metabolome-wide association study showed that 128 metabolites were significantly altered in the ligated mice compared with the sham group. Of these, sphingomyelin (SM; m/z 703.5747), a common mammalian cell membrane sphingolipid, was most significantly increased in the ligated mice. Of the 128 discriminatory metabolites, 18 and 41 were positively and negatively correlated with SM, respectively. The amino acids methionine and phenylalanine were increased by d-flow, while phosphatidylcholine and phosphatidylethanolamine were decreased by d-flow, and these metabolites were correlated with SM. Other significantly affected metabolites included dietary and environmental agents. Pathway analysis showed that the metabolic changes of d-flow impacted broad functional networks. These results suggest that signaling from d-flow occurring in focal regions induces systemic metabolic changes associated with atherosclerosis.
Collapse
Affiliation(s)
- Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Chan Woo Kim
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Douglas I Walker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia; Department of Civil and Environmental Engineering, Tufts University, Medford, Massachusetts; and
| | - Dong Won Kang
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Sandeep Kumar
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Michael Orr
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Karan Uppal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia
| | - Hanjoong Jo
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia;
| |
Collapse
|
42
|
Lou B, Dong J, Li Y, Ding T, Bi T, Li Y, Deng X, Ye D, Jiang XC. Pharmacologic inhibition of sphingomyelin synthase (SMS) activity reduces apolipoprotein-B secretion from hepatocytes and attenuates endotoxin-mediated macrophage inflammation. PLoS One 2014; 9:e102641. [PMID: 25032960 PMCID: PMC4102531 DOI: 10.1371/journal.pone.0102641] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 06/20/2014] [Indexed: 01/06/2023] Open
Abstract
Sphingomyelin synthase (SMS) plays an important role in plasma atherogenic lipoprotein metabolism, inflammation, and the development of atherosclerosis. To understand whether the impaired apoB secretion and inflammation response is a direct result from lack of SMS activity, in this study, we prepared a series of compounds that inhibit SMS activity. Further, we characterized Dy105, the most potent inhibitor. We found that Dy105 treatment significantly reduces SM levels in SM-rich microdomain on cell membranes. Moreover, we found that SMS inhibition reduces apoB secretion in a human hepatoma cell line and reduces the activation of NFκB and p38, a MAP kinase, in bone marrow derived macrophages. These studies provided further evidence that SMS activity regulates atherogenic lipoprotein metabolism and inflammatory responses. Pharmacologic inhibition of SMS may be a new therapy for atherosclerosis by reducing apoB secretion, and reducing inflammation.
Collapse
Affiliation(s)
- Bin Lou
- School of Pharmacy Fudan University, Shanghai, China
- * E-mail: (BL); (DY)
| | - Jibin Dong
- School of Pharmacy Fudan University, Shanghai, China
| | - Yali Li
- School of Pharmacy Fudan University, Shanghai, China
| | - Tingbo Ding
- School of Pharmacy Fudan University, Shanghai, China
| | - Tingting Bi
- School of Pharmacy Fudan University, Shanghai, China
| | - Yue Li
- School of Pharmacy Fudan University, Shanghai, China
| | - Xiaodong Deng
- School of Pharmacy Fudan University, Shanghai, China
| | - Deyong Ye
- School of Pharmacy Fudan University, Shanghai, China
- * E-mail: (BL); (DY)
| | - Xian-Cheng Jiang
- School of Pharmacy Fudan University, Shanghai, China
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York United States of America
| |
Collapse
|
43
|
Taniguchi M, Okazaki T. The role of sphingomyelin and sphingomyelin synthases in cell death, proliferation and migration—from cell and animal models to human disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:692-703. [DOI: 10.1016/j.bbalip.2013.12.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 12/06/2013] [Accepted: 12/09/2013] [Indexed: 12/16/2022]
|
44
|
Endogenous ceramide contributes to the transcytosis of oxLDL across endothelial cells and promotes its subendothelial retention in vascular wall. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:823071. [PMID: 24817993 PMCID: PMC4003761 DOI: 10.1155/2014/823071] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/17/2014] [Accepted: 03/21/2014] [Indexed: 11/24/2022]
Abstract
Oxidized low density of lipoprotein (oxLDL) is the major lipid found in atherosclerotic lesion and elevated plasma oxLDL is recognized to be a risk factor of atherosclerosis. Whether plasma oxLDL could be transported across endothelial cells and initiate atherosclerotic changes remains unknown. In an established in vitro cellular transcytosis model, the present study found that oxLDL could traffic across vascular endothelial cells and further that the regulation of endogenous ceramide production by ceramide metabolizing enzyme inhibitors significantly altered the transcytosis of oxLDL across endothelial cells. It was found that acid sphingomyelinase inhibitor, desipramine (DES), and de novo ceramide synthesis inhibitor, myriocin (MYR), both decreasing the endogenous ceramide production, significantly inhibited the transcytosis of oxLDL. Ceramidase inhibitor, N-oleoylethanolamine (NOE), and sphingomyelin synthase inhibitor, O-Tricyclo[5.2.1.02,6]dec-9-yl dithiocarbonate potassium salt (D609), both increasing the endogenous ceramide production, significantly upregulated the transcytosis of oxLDL. In vivo, injection of fluorescence labeled oxLDL into mice body also predisposed to the subendothelial retention of these oxidized lipids. The observations provided in the present study demonstrate that endogenous ceramide contributes to the transcytosis of oxLDL across endothelial cells and promotes the initiating step of atherosclerosis—the subendothelial retention of lipids in vascular wall.
Collapse
|
45
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
46
|
Lietz M, Berges A, Lebrun S, Meurrens K, Steffen Y, Stolle K, Schueller J, Boue S, Vuillaume G, Vanscheeuwijck P, Moehring M, Schlage W, De Leon H, Hoeng J, Peitsch M. Cigarette-smoke-induced atherogenic lipid profiles in plasma and vascular tissue of apolipoprotein E-deficient mice are attenuated by smoking cessation. Atherosclerosis 2013; 229:86-93. [DOI: 10.1016/j.atherosclerosis.2013.03.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 02/28/2013] [Accepted: 03/26/2013] [Indexed: 11/16/2022]
|
47
|
Hornemann T, Worgall TS. Sphingolipids and atherosclerosis. Atherosclerosis 2012; 226:16-28. [PMID: 23075523 DOI: 10.1016/j.atherosclerosis.2012.08.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 08/06/2012] [Accepted: 08/22/2012] [Indexed: 11/19/2022]
Abstract
The atherosclerotic lesion contains a high amount of sphingolipids, a large group of structurally diverse lipids that regulate distinct biological functions beyond their role as structural membrane components. Assessment of their role in atherogenesis has been enabled after genes that regulate their metabolism had been identified and facilitated by the more wide availability of mass spectrometry. Here we discuss recent mechanistic insights obtained in animal and epidemiological studies that have greatly enhanced our understanding of mechanisms how sphingolipids affect the atherosclerotic process.
Collapse
Affiliation(s)
- Thorsten Hornemann
- Inst. for Clinical Chemistry, University Hospital Zuerich, Raemistrasse 100, 8091 Zuerich, Switzerland.
| | | |
Collapse
|
48
|
Yazdanyar A, Jiang XC. Liver phospholipid transfer protein (PLTP) expression with a PLTP-null background promotes very low-density lipoprotein production in mice. Hepatology 2012; 56:576-84. [PMID: 22367708 PMCID: PMC3409695 DOI: 10.1002/hep.25648] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 01/31/2012] [Indexed: 12/07/2022]
Abstract
UNLABELLED It is known that plasma phospholipid transfer protein (PLTP) activity influences lipoprotein metabolism. The liver is one of the major sites of lipoprotein production and degradation, as well as of PLTP expression. To address the impact of liver-expressed PLTP on lipoprotein metabolism, we created a mouse model that expresses PLTP in the liver acutely and specifically, with a PLTP-null background. This approach in mouse model preparations can also be used universally for evaluating the function of many other genes in the liver. We found that liver PLTP expression dramatically increases plasma levels of non-high-density lipoprotein (HDL) cholesterol (2.7-fold, P < 0.0001), non-HDL phospholipid (2.5-fold, P < 0.001), and triglyceride (51%, P < 0.01), but has no significant influence on plasma HDL lipids compared with controls. Plasma apolipoprotein (apo)B levels were also significantly increased in PLTP-expressing mice (2.2-fold, P < 0.001), but those of apoA-I were not. To explore the mechanism involved, we examined the lipidation and secretion of nascent very low-density lipoprotein (VLDL), finding that liver PLTP expression significantly increases VLDL lipidation in hepatocyte microsomal lumina, and also VLDL secretion into the plasma. CONCLUSION It is possible to prepare a mouse model that expresses the gene of interest only in the liver, but not in other tissues. Our results suggest, for the first time, that the major function of liver PLTP is to drive VLDL production and makes a small contribution to plasma PLTP activity.
Collapse
Affiliation(s)
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center
- To whom correspondence may be addressed at Downstate Medical Center, 450 Clarkson Ave. Box 5 Brooklyn, NY 11203, tel. (718) 270-6701, Fax (718) 270-3732,
| |
Collapse
|
49
|
Li Z, Fan Y, Liu J, Li Y, Huan C, Bui HH, Kuo MS, Park TS, Cao G, Jiang XC. Impact of sphingomyelin synthase 1 deficiency on sphingolipid metabolism and atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2012; 32:1577-84. [PMID: 22580896 DOI: 10.1161/atvbaha.112.251538] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Sphingomyelin synthase (SMS) catalyzes the conversion of ceramide to sphingomyelin and sits at the crossroads of sphingolipid biosynthesis. SMS has 2 isoforms: SMS1 and SMS2. Although they have the same SMS activity, they are different enzymes with distinguishable subcellular localizations and cell expression patterns. It is conceivable that these differences could yield different consequences, in terms of sphingolipid metabolism and its related atherogenesis. METHODS AND RESULTS We created Sms1 gene knockout mice and found that Sms1 deficiency significantly decreased plasma, liver, and macrophage sphingomyelin (59%, 45%, and 54%, respectively), but only had a marginal effect on ceramide levels. Surprisingly, we found that Sms1 deficiency dramatically increased glucosylceramide and GM3 levels in plasma, liver, and macrophages (4- to 12-fold), whereas Sms2 deficiency had no such effect. We evaluated the total SMS activity in tissues and found that Sms1 deficiency causes 77% reduction in SMS activity in macrophages, indicating SMS1 is the major SMS in macrophages. Moreover, Sms1-deficient macrophages have a significantly higher glucosylceramide synthase activity. We also found that Sms1 deficiency significantly attenuated toll-like 4 receptor-mediated nuclear factor-κB and mitogen-activated protein kinase activation after lipopolysaccharide treatment. To evaluate atherogenicity, we transplanted Sms1 knockout mouse bone marrow into low-density lipoprotein receptor knockout mice (Sms1(-/-)→Ldlr(-/-)). After 3 months on a western diet, these animals showed a significant decrease of atherosclerotic lesions in the root and the entire aorta (35% and 44%, P<0.01, respectively) and macrophage content in lesions (51%, P<0.05), compared with wild-type→Ldlr(-/-) mice. CONCLUSIONS Sms1 deficiency decreases sphingomyelin, but dramatically increases the levels of glycosphingolipids. Atherosclerosis in Sms1(-/-)→Ldlr(-/-) mice is significantly decreased.
Collapse
Affiliation(s)
- Zhiqiang Li
- Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Subbaiah PV, Jiang XC, Belikova NA, Aizezi B, Huang ZH, Reardon CA. Regulation of plasma cholesterol esterification by sphingomyelin: effect of physiological variations of plasma sphingomyelin on lecithin-cholesterol acyltransferase activity. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:908-13. [PMID: 22370449 DOI: 10.1016/j.bbalip.2012.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 02/06/2012] [Accepted: 02/10/2012] [Indexed: 01/22/2023]
Abstract
Although sphingomyelin (SM) is the most abundant phospholipid in the plasma, next to phosphatidylcholine (PC), its physiological function in plasma is unclear. Here we employed plasma from various genetic models of mice which naturally differ in their plasma SM/PC ratios, to study the role of SM as a modulator of LCAT, the enzyme responsible for HDL maturation and the synthesis of cholesteryl esters (CE) in normal plasma. Serine palmitoyltransferase deficient mice, and SM synthase deficient mice, both of which have below normal SM/PC ratios, showed significantly elevated LCAT activities when assayed with the endogenous substrates. On the other hand, LDL receptor knockout mice, and apo E knockout mice, both of which have high SM/PC ratios, had markedly reduced (-80%) LCAT activities. The LCAT levels in plasma, as assayed with an exogenous substrate, were similar in all groups, except for a 45% decrease in apo E knockout mice. Plasma samples with high SM/PC ratios had lower percentage of 20:4, 22:5, and 22:6 CE all of which are formed by LCAT, and a higher percentage of the atherogenic 18:1 CE which is mainly derived from the action of liver ACAT, showing that in vivo, the contribution of LCAT to plasma CE is reduced while that of liver ACAT is increased. These results show that SM is a physiological modulator of LCAT activity as well as plasma CE composition, and this may contribute to the previously reported pro-atherogenic effect of high plasma SM levels.
Collapse
|