1
|
Chen J, Zhao D, Wang Y, Liu M, Zhang Y, Feng T, Xiao C, Song H, Miao R, Xu L, Chen H, Qiu X, Xu Y, Xu J, Cui Z, Wang W, Quan Y, Zhu Y, Huang C, Zheng SG, Zhao J, Zhu T, Sun L, Fan G. Lactylated Apolipoprotein C-II Induces Immunotherapy Resistance by Promoting Extracellular Lipolysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406333. [PMID: 38981044 PMCID: PMC11481198 DOI: 10.1002/advs.202406333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Indexed: 07/11/2024]
Abstract
Mortality rates due to lung cancer are high worldwide. Although PD-1 and PD-L1 immune checkpoint inhibitors boost the survival of patients with non-small-cell lung cancer (NSCLC), resistance often arises. The Warburg Effect, which causes lactate build-up and potential lysine-lactylation (Kla), links immune dysfunction to tumor metabolism. The role of non-histone Kla in tumor immune microenvironment and immunotherapy remains to be clarified. Here, global lactylome profiling and metabolomic analyses of samples from patients with NSCLC is conducted. By combining multi-omics analysis with in vitro and in vivo validation, that intracellular lactate promotes extracellular lipolysis through lactyl-APOC2 is revealed. Mechanistically, lactate enhances APOC2 lactylation at K70, stabilizing it and resulting in FFA release, regulatory T cell accumulation, immunotherapy resistance, and metastasis. Moreover, the anti-APOC2K70-lac antibody that sensitized anti-PD-1 therapy in vivo is developed. This findings highlight the potential of anti lactyl-APOC2-K70 approach as a new combination therapy for sensitizing immunotherapeutic responses.
Collapse
Affiliation(s)
- Jian Chen
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University507 Zhengmin RoadShanghai200433P. R. China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University507 Zhengmin RoadShanghai200433P. R. China
| | - Yupeng Wang
- Department of General Surgery, Shanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011P. R. China
| | - Ming Liu
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University507 Zhengmin RoadShanghai200433P. R. China
| | - Yuan Zhang
- Department of Gastrointestinal SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Tingting Feng
- Department of Clinical PharmacyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Chao Xiao
- Department of Gastrointestinal SurgeryShanghai East Hospital, School of MedicineTongji UniversityShanghai200040P. R. China
| | - Huan Song
- Department of Clinical Laboratory MedicineShanghai Pulmonary HospitalTongji University School of MedicineShanghai200433P. R. China
| | - Rui Miao
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Li Xu
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University507 Zhengmin RoadShanghai200433P. R. China
| | - Hongwei Chen
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Xiaoying Qiu
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Yi Xu
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Jingxuan Xu
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty MannheimHeidelberg University68167MannheimGermany
| | - Zelin Cui
- Department of Laboratory MedicineShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Wei Wang
- Department of Breast‐thyroid SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Yanchun Quan
- Central LaboratoryLinyi People's HospitalShandong273300P. R. China
| | - Yifeng Zhu
- Department of Internal Medicine II, Klinikum rechts der IsarTechnical University of MunichIsmaninger Str. 2281675MunichGermany
| | - Chen Huang
- Department of Gastrointestinal SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research InstituteShanghai Jiaotong University School of Medicine Affiliated Songjiang HospitalShanghai200080P. R. China
| | - Jian‐yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092P. R. China
| | - Ting Zhu
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Lianhui Sun
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092P. R. China
| | - Guangjian Fan
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| |
Collapse
|
2
|
Cofiño-Fabres C, Boonen T, Rivera-Arbeláez JM, Rijpkema M, Blauw L, Rensen PCN, Schwach V, Ribeiro MC, Passier R. Micro-Engineered Heart Tissues On-Chip with Heterotypic Cell Composition Display Self-Organization and Improved Cardiac Function. Adv Healthc Mater 2024; 13:e2303664. [PMID: 38471185 DOI: 10.1002/adhm.202303664] [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: 10/26/2023] [Revised: 01/30/2024] [Indexed: 03/14/2024]
Abstract
Advanced in vitro models that recapitulate the structural organization and function of the human heart are highly needed for accurate disease modeling, more predictable drug screening, and safety pharmacology. Conventional 3D Engineered Heart Tissues (EHTs) lack heterotypic cell complexity and culture under flow, whereas microfluidic Heart-on-Chip (HoC) models in general lack the 3D configuration and accurate contractile readouts. In this study, an innovative and user-friendly HoC model is developed to overcome these limitations, by culturing human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs), endothelial (ECs)- and smooth muscle cells (SMCs), together with human cardiac fibroblasts (FBs), underflow, leading to self-organized miniaturized micro-EHTs (µEHTs) with a CM-EC interface reminiscent of the physiological capillary lining. µEHTs cultured under flow display enhanced contractile performance and conduction velocity. In addition, the presence of the EC layer altered drug responses in µEHT contraction. This observation suggests a potential barrier-like function of ECs, which may affect the availability of drugs to the CMs. These cardiac models with increased physiological complexity, will pave the way to screen for therapeutic targets and predict drug efficacy.
Collapse
Affiliation(s)
- Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Tom Boonen
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
| | - José M Rivera-Arbeláez
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- BIOS Lab-on-a-Chip Group, MESA+ Institute for Nanotechnology, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Minke Rijpkema
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Lisanne Blauw
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Marcelo C Ribeiro
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, 2300 RC, The Netherlands
| |
Collapse
|
3
|
Papadopoulou P, van der Pol R, van Hilten N, van Os WL, Pattipeiluhu R, Arias-Alpizar G, Knol RA, Noteborn W, Moradi MA, Ferraz MJ, Aerts JMFG, Sommerdijk N, Campbell F, Risselada HJ, Sevink GJA, Kros A. Phase-Separated Lipid-Based Nanoparticles: Selective Behavior at the Nano-Bio Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310872. [PMID: 37988682 DOI: 10.1002/adma.202310872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Indexed: 11/23/2023]
Abstract
The membrane-protein interface on lipid-based nanoparticles influences their in vivo behavior. Better understanding may evolve current drug delivery methods toward effective targeted nanomedicine. Previously, the cell-selective accumulation of a liposome formulation in vivo is demonstrated, through the recognition of lipid phase-separation by triglyceride lipases. This exemplified how liposome morphology and composition can determine nanoparticle-protein interactions. Here, the lipase-induced compositional and morphological changes of phase-separated liposomes-which bear a lipid droplet in their bilayer- are investigated, and the mechanism upon which lipases recognize and bind to the particles is unravelled. The selective lipolytic degradation of the phase-separated lipid droplet is observed, while nanoparticle integrity remains intact. Next, the Tryptophan-rich loop of the lipase is identified as the region with which the enzymes bind to the particles. This preferential binding is due to lipid packing defects induced on the liposome surface by phase separation. In parallel, the existing knowledge that phase separation leads to in vivo selectivity, is utilized to generate phase-separated mRNA-LNPs that target cell-subsets in zebrafish embryos, with subsequent mRNA delivery and protein expression. Together, these findings can expand the current knowledge on selective nanoparticle-protein communications and in vivo behavior, aspects that will assist to gain control of lipid-based nanoparticles.
Collapse
Affiliation(s)
- Panagiota Papadopoulou
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Rianne van der Pol
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Niek van Hilten
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Winant L van Os
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Roy Pattipeiluhu
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Gabriela Arias-Alpizar
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Renzo Aron Knol
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Willem Noteborn
- NeCEN, Leiden University, Einsteinweg 55, Leiden, 2333 AL, The Netherlands
| | - Mohammad-Amin Moradi
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P. O. Box 513, Eindhoven, 5600 MB, The Netherlands
| | - Maria Joao Ferraz
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | | | - Nico Sommerdijk
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P. O. Box 513, Eindhoven, 5600 MB, The Netherlands
- Department of Medical BioSciences and Radboud Technology Center - Electron Microscopy, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Herre Jelger Risselada
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
- Department of Physics, Technical University Dortmund, 44221, Dortmund, Germany
| | - Geert Jan Agur Sevink
- Department of Biophysical Organic Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| |
Collapse
|
4
|
Bavis MM, Nicholas AM, Tobin AJ, Christian SL, Brown RJ. The breast cancer microenvironment and lipoprotein lipase: Another negative notch for a beneficial enzyme? FEBS Open Bio 2023; 13:586-596. [PMID: 36652113 PMCID: PMC10068309 DOI: 10.1002/2211-5463.13559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/20/2022] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
The energy demand of breast cancers is in part met through the β-oxidation of exogenous fatty acids. Fatty acids may also be used to aid in cell signaling and toward the construction of new membranes for rapidly proliferating tumor cells. A significant quantity of fatty acids comes from the hydrolysis of lipoprotein triacylglycerols and phospholipids by lipoprotein lipase (LPL). The lipid obtained via LPL in the breast tumor microenvironment may thus promote breast tumor growth and development. In this hypothesis article, we introduce LPL, provide a meta-analysis of RNAseq data showing that LPL is associated with poor prognosis, and explain how LPL might play a role in breast cancer prognosis over time.
Collapse
Affiliation(s)
- Makayla M Bavis
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Allison M Nicholas
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Alexandria J Tobin
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Sherri L Christian
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Robert J Brown
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
5
|
Li L, Xie W. LncRNA HDAC11-AS1 Suppresses Atherosclerosis by Inhibiting HDAC11-Mediated Adropin Histone Deacetylation. J Cardiovasc Transl Res 2022; 15:1256-1269. [PMID: 35505157 DOI: 10.1007/s12265-022-10248-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/31/2022] [Indexed: 12/16/2022]
Abstract
LncRNA HDAC11-AS1 (HDAC11-AS1) is the natural antisense transcript of HDAC11, a key enzyme for DNA histone deacetylation. We evaluated the role of HDAC11-AS1 in atherosclerosis. In this research, we found that HDAC11-AS1 ameliorated blood lipid levels and atherosclerosis in high fat-dieted apoE-/- mice by regulating HDAC11 negatively. The change in blood lipid levels is related to the expression of LPL, which is enhanced by HDAC11-AS1 through regulating adropin histone deacetylation in vitro and in vivo. In conclusion, HDAC11-AS1 plays an anti-atherogenic role through adropin to induce LPL expressions, thereby enhancing TG metabolism. The results are valuable for the further development of HDAC11-AS1 and its clinical applications. It provides a new clinical therapeutic target for cardiovascular disease treatment.
Collapse
Affiliation(s)
- Liang Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
6
|
Zhang B, Dong C, Li S, Song X, Wei W, Liu L. Triglyceride to High-Density Lipoprotein Cholesterol Ratio is an Important Determinant of Cardiovascular Risk and Poor Prognosis in Coronavirus Disease-19: A Retrospective Case Series Study. Diabetes Metab Syndr Obes 2020; 13:3925-3936. [PMID: 33122929 PMCID: PMC7591232 DOI: 10.2147/dmso.s268992] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/26/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Acute myocardial injury and heart failure characterized by elevated cardiac troponin and decreased heart pump function are significant clinical features and prognostic factors of coronavirus disease-19 (COVID-19). Triglyceride to high-density lipoprotein cholesterol (TG/HDL-C) ratio is an indicator of insulin resistance. This study aimed to explore the association of the TG/HDL-C ratio with cardiovascular risk and prognosis in COVID-19. METHODS Ninety-eight laboratory-confirmed patients with COVID-19 admitted in a tertiary teaching hospital in Wuhan, China, were enrolled in this retrospective study. Regression models were used to investigate the association between TG/HDL-C ratio with myocardial injury, heart failure, severity, and mortality in COVID-19. RESULTS Among the 98 patients, the mean age was 63.9±1.4 years, and male sex (58, 59%) was predominant. Forty-six patients (47%) were admitted to the intensive care unit (ICU), 32 (33%) and 46 (47%) patients suffered from myocardial injury and heart failure, respectively, and 36 (37%) patients died. The TG/HDL-C ratio was increased in patients with myocardial injury, heart failure, severe illness, and fatal outcome (P<0.05 for each). Baseline TG/HDL-C ratio significantly correlated with log transformed levels of plasma high-sensitivity cardiac troponin I (r=0.251, P=0.018), N-terminal brain natriuretic propeptide (r=0.274, P=0.008), glycated hemoglobin (r=0.239, P=0.038), and interleukin-6 (r=0.218, P=0.042). Multivariate logistic regression analysis showed that an increased TG/HDL-C ratio was independently associated with the risk of myocardial injury [odds ratio (OR)=2.73; P=0.013], heart failure (OR=2.64; P=0.019), disease severity (OR=3.01; P=0.032), and fatal outcome (OR=2.97; P=0.014). CONCLUSION Increased TG/HDL-C ratio was independently associated with myocardial injury, heart failure, disease severity, and mortality in patients with COVID-19, and it may be a useful marker for early identification of patients with high risk and poor outcome.
Collapse
Affiliation(s)
- Benping Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei430030, People’s Republic of China
| | - Chen Dong
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei430030, People’s Republic of China
| | - Shengzhong Li
- Department of Surgery, Wuhan Jinyintan Hospital, Wuhan, Hubei430023, People’s Republic of China
| | - Xiaoqing Song
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei430030, People’s Republic of China
| | - Wang Wei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei430030, People’s Republic of China
| | - Li Liu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei430030, People’s Republic of China
- Correspondence: Li Liu Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, Hubei430030, People’s Republic of ChinaTel +86-027-83663470Fax +86-027-83662883 Email
| |
Collapse
|
7
|
Colliva A, Braga L, Giacca M, Zacchigna S. Endothelial cell-cardiomyocyte crosstalk in heart development and disease. J Physiol 2019; 598:2923-2939. [PMID: 30816576 PMCID: PMC7496632 DOI: 10.1113/jp276758] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/29/2019] [Indexed: 12/15/2022] Open
Abstract
The crosstalk between endothelial cells and cardiomyocytes has emerged as a requisite for normal cardiac development, but also a key pathogenic player during the onset and progression of cardiac disease. Endothelial cells and cardiomyocytes are in close proximity and communicate through the secretion of paracrine signals, as well as through direct cell-to-cell contact. Here, we provide an overview of the endothelial cell-cardiomyocyte interactions controlling heart development and the main processes affecting the heart in normal and pathological conditions, including ischaemia, remodelling and metabolic dysfunction. We also discuss the possible role of these interactions in cardiac regeneration and encourage the further improvement of in vitro models able to reproduce the complex environment of the cardiac tissue, in order to better define the mechanisms by which endothelial cells and cardiomyocytes interact with a final aim of developing novel therapeutic opportunities.
Collapse
Affiliation(s)
- Andrea Colliva
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 34149, Trieste, Italy
| | - Luca Braga
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 34149, Trieste, Italy
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 34149, Trieste, Italy.,Biotechnology Development Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 34149, Trieste, Italy
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 34149, Trieste, Italy.,Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, 34149, Trieste, Italy
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Lipoprotein lipase (LpL) is well known for its lipolytic action in blood lipoprotein triglyceride catabolism. This article summarizes the recent mechanistic and molecular studies on elucidating the 'unconventional' roles of LpL in mediating biological events related to immune cell response and lipid transport in the pathogenesis of cardiovascular disease (CVD) and tissue degenerative disorders. RECENT FINDINGS Several approaches to inactivate the inhibitors that block LpL enzymatic activity have reestablished the importance of systemic LpL activity in reducing CVD risk. On the other hand, increasing evidence suggests that focal arterial expression of LpL relates to aortic macrophage levels and inflammatory processes. In the hematopoietic origin, LpL also plays a role in modulating hematopoietic stem cell proliferation and circulating blood cell levels and phenotypes. Finally, building upon the strong genetic evidence on the association with assorted brain disorders, a new era in exploring the mechanistic insights into the functions and activity of LpL in brain that impacts central nerve systems has begun. SUMMARY A better understanding of the molecular action of LpL will help to devise novel strategies for intervention of a number of diseases, including blood cell or metabolic disorders, as well to inhibit pathways related to CVD and tissue degenerative processes.
Collapse
Affiliation(s)
- Chuchun L Chang
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
9
|
Xue Z, Zhang Q, Yu W, Wen H, Hou X, Li D, Kou X. Potential Lipid-Lowering Mechanisms of Biochanin A. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3842-3850. [PMID: 28447802 DOI: 10.1021/acs.jafc.7b00967] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Extensive studies have demonstrated that biochanin A (BCA) has a significant hypolipidemic effect. However, its mechanism of action is not clear. In this context, the effect of BCA on a high-fat diet (HFD)-induced hyperlipidemia in mice was determined. The results showed that treatment with a medium dose of biochanin A (BM) significantly decreased low-density lipoprotein cholesterol (LDL-C) 85% (from 1.196 ± 0.183 to 0.181 ± 0.0778 mM) and total cholesterol (TC) 39% (from 5.983 ± 0.128 to 3.649 ± 0.374 mM) levels, increased lipoprotein lipase (LPL) 96% (from 1.421 ± 0.0982 to 2.784 ± 0.177 U/mg protein) and hepatic triglyceride lipase (HTGL) 78% (from 1.614 ± 0.0848 to 2.870 ± 0.0977 U/mg protein) activities, significantly improved fecal lipid levels, and lowered the epididymal fat index in hyperlipidemic mice compared with the HFD control mice (p < 0.05). In vitro, the high antioxidant capacity of BCA was determined by the FRAP assay, ABTS•+ scavenging method, and an ROS assay. In RAW 264.7 macrophages, a dose of 10 μM BCA significantly increased the cholesterol efflux by 18.7% compared with the control cells. Moreover, molecular docking of BCA on cholesterol ester transfer protein (CETP) (Asn24 and Thr27 at the N-terminal; Ala274 and Phe270 at the C-terminal) gave new insights into the role of BCA in preventing cholesterol ester transport.
Collapse
Affiliation(s)
- Zhaohui Xue
- School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, China
| | - Qian Zhang
- School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, China
| | - Wancong Yu
- Medical Plant Laboratory, Tianjin Research Center of Agricultural Biotechnology , Tianjin 300381, China
| | - Haichao Wen
- School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, China
| | - Xiaonan Hou
- School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, China
| | - Dan Li
- School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, China
| | - Xiaohong Kou
- School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, China
| |
Collapse
|
10
|
Blanchard PG, Turcotte V, Côté M, Gélinas Y, Nilsson S, Olivecrona G, Deshaies Y, Festuccia WT. Peroxisome proliferator-activated receptor γ activation favours selective subcutaneous lipid deposition by coordinately regulating lipoprotein lipase modulators, fatty acid transporters and lipogenic enzymes. Acta Physiol (Oxf) 2016; 217:227-39. [PMID: 26918671 DOI: 10.1111/apha.12665] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/15/2015] [Accepted: 02/19/2016] [Indexed: 12/18/2022]
Abstract
AIM Peroxisome proliferator-activated receptor (PPAR) γ activation is associated with preferential lipoprotein lipase (LPL)-mediated fatty acid storage in peripheral subcutaneous fat depots. How PPARγ agonism acts upon the multi-level modulation of depot-specific lipid storage remains incompletely understood. METHODS We evaluated herein triglyceride-derived lipid incorporation into adipose tissue depots, LPL mass and activity, mRNA levels and content of proteins involved in the modulation of LPL activity and fatty acid transport, and the expression/activity of enzymes defining adipose tissue lipogenic potential in rats treated with the PPARγ ligand rosiglitazone (30 mg kg(-1) day(-1) , 23 days) after either a 10-h fasting period or a 17-h fast followed by 6 h of ad libitum refeeding. RESULTS Rosiglitazone stimulated lipid accretion in subcutaneous fat (SF) ~twofold and significantly reduced that of visceral fat (VF) to nearly half. PPARγ activation selectively increased LPL mass, activity and the expression of its chaperone LMF1 in SF. In VF, rosiglitazone had no effect on LPL activity and downregulated the mRNA levels of the transendothelial transporter GPIHBP1. Overexpression of lipid uptake and fatty acid transport proteins (FAT/CD36, FATP1 and FABP4) and stimulation of lipogenic enzyme activities (GPAT, AGPAT and DGAT) upon rosiglitazone treatment were of higher magnitude in SF. CONCLUSIONS Together these findings demonstrate that the depot-specific transcriptional control of LPL induced by PPARγ activation extends to its key interacting proteins and post-translational modulators to favour subcutaneous lipid storage.
Collapse
Affiliation(s)
- P. G. Blanchard
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - V. Turcotte
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - M. Côté
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - Y. Gélinas
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - S. Nilsson
- Department of Medical Biosciences/Physiological Chemistry; Umeå University; Umeå Sweden
| | - G. Olivecrona
- Department of Medical Biosciences/Physiological Chemistry; Umeå University; Umeå Sweden
| | - Y. Deshaies
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - W. T. Festuccia
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| |
Collapse
|
11
|
Zhang L, Wang HH. The essential functions of endoplasmic reticulum chaperones in hepatic lipid metabolism. Dig Liver Dis 2016; 48:709-16. [PMID: 27133206 DOI: 10.1016/j.dld.2016.03.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 03/06/2016] [Accepted: 03/22/2016] [Indexed: 12/11/2022]
Abstract
The endoplasmic reticulum (ER) is an essential organelle for protein and lipid synthesis in hepatocytes. ER homeostasis is vital to maintain normal hepatocyte physiology. Perturbed ER functions causes ER stress associated with accumulation of unfolded protein in the ER that activates a series of adaptive signalling pathways, termed unfolded protein response (UPR). The UPR regulates ER chaperone levels to preserve ER protein-folding environment to protect the cell from ER stress. Recent findings reveal an array of ER chaperones that alter the protein-folding environment in the ER of hepatocytes and contribute to dysregulation of hepatocyte lipid metabolism and liver disease. In this review, we will discuss the specific functions of these chaperones in regulation of lipid metabolism, especially de novo lipogenesis and lipid transport and demonstrate their homeostatic role not only for ER-protein synthesis but also for lipid metabolism in hepatocyte.
Collapse
Affiliation(s)
- LiChun Zhang
- Department of Emergency, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Hong-Hui Wang
- College of Biology, Hunan University, Changsha, Hunan Province, China.
| |
Collapse
|
12
|
Peng J, Lv YC, He PP, Tang YY, Xie W, Liu XY, Li Y, Lan G, Zhang M, Zhang C, Shi JF, Zheng XL, Yin WD, Tang CK. RETRACTED:Betulinic acid downregulates expression of oxidative stress-induced lipoprotein lipase via the PKC/ERK/c-Fos pathway in RAW264.7 macrophages. Biochimie 2015; 119:192-203. [PMID: 26542288 DOI: 10.1016/j.biochi.2015.10.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 10/28/2015] [Indexed: 12/22/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. Concerns raised by Dr. Sander Kersten in PubPeer pointed out that Figures 6.1B and 6.2B of this paper were different figures but the legends and Western blots were identical; the quantification was also seen to be different between the two figures. Shortly afterwards, the authors asked to publish a corrigendum for part B of Figure 6.1, including images of western blots and associated bar plots. Subsequently, the journal conducted an investigation and found evidence that there had been improper manipulation and duplication of images in Figures 2 E, 6.2 B, 5 A and and 6.2 D, as shown by the reuse of several western blot bands with approximately 180° rotation in each case. After raising the complaint with the authors, the corresponding author agreed that the paper should be retracted. The authors apologise to the readers of the journal.
Collapse
Affiliation(s)
- Juan Peng
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China; Yongzhou Vocational and Technical College, Yongzhou, Hunan 425000, China
| | - Yun-Cheng Lv
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Ping-Ping He
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China; School of Nursing, University of South China, Hengyang, Hunan 421001, China
| | - Yan-Yan Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Wei Xie
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Xiang-Yu Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Technology, University of South, Hengyang, Hunan 421001, China
| | - Yuan Li
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Gan Lan
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Min Zhang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Chi Zhang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Jin-Feng Shi
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Wei-Dong Yin
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
13
|
Borén J, Watts GF, Adiels M, Söderlund S, Chan DC, Hakkarainen A, Lundbom N, Matikainen N, Kahri J, Vergès B, Barrett PHR, Taskinen MR. Kinetic and Related Determinants of Plasma Triglyceride Concentration in Abdominal Obesity: Multicenter Tracer Kinetic Study. Arterioscler Thromb Vasc Biol 2015; 35:2218-24. [PMID: 26315407 DOI: 10.1161/atvbaha.115.305614] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 08/04/2015] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Patients with obesity and diabetes mellitus have increased risk of cardiovascular disease. A major cause is an atherogenic dyslipidemia related primarily to elevated plasma concentrations of triglyceride-rich lipoproteins. The aim of this study was to clarify determinants of plasma triglyceride concentration. We focused on factors that predict the kinetics of very-low density lipoprotein 1 (VLDL1) triglycerides. APPROACH AND RESULTS A multicenter study using dual stable isotopes (deuterated leucine and glycerol) and multicompartmental modeling was performed to elucidate the kinetics of triglycerides and apoB in VLDL1 in 46 subjects with abdominal obesity and additional cardiometabolic risk factors. Results showed that plasma triglyceride concentrations were dependent on both the secretion rate (r=0.44, P<0.01; r=0.45, P<0.01) and fractional catabolism (r=0.49, P<0.001; r=0.55, P<0.001) of VLDL1-triglycerides and VLDL1-apoB. Liver fat mass was independently and directly associated with secretion rates of VLDL1-triglycerides (r=0.56, P<0.001) and VLDL1-apoB (r=0.53, P<0.001). Plasma apoC-III concentration was independently and inversely associated with the fractional catabolisms of VLDL1-triglycerides (r=0.48, P<0.001) and VLDL1-apoB (r=0.51, P<0.001). CONCLUSIONS Plasma triglyceride concentrations in abdominal obesity are determined by the kinetics of VLDL1 subspecies, catabolism being mainly dependent on apoC-III concentration and secretion on liver fat content. Reduction in liver fat and targeting apoC-III may be an effective approach for correcting triglyceride metabolism atherogenic dyslipidemia in obesity.
Collapse
Affiliation(s)
- Jan Borén
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.).
| | - Gerald F Watts
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Martin Adiels
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Sanni Söderlund
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Dick C Chan
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Antti Hakkarainen
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Nina Lundbom
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Niina Matikainen
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Juhani Kahri
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Bruno Vergès
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - P Hugh R Barrett
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| | - Marja-Riitta Taskinen
- From the Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden (J.B., M.A.); Lipid Disorders Clinic, Metabolic Research Centre, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology (G.F.W., D.C.C., P.H.R.B.) and Faculty of Engineering, Computing and Mathematics (P.H.R.B.), University of Western Australia, Perth, Australia; Heart and Lung Centre, Helsinki University Central Hospital and Research Programs' Unit, Diabetes and Obesity (S.S., N.M., M.-R.T.) and Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital (A.H., N.L.), University of Helsinki, Helsinki, Finland; Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland (N.M., J.K.); and Department of Endocrinology-Diabetology, University Hospital and INSERM CRI 866, Dijon, France (B.V.)
| |
Collapse
|
14
|
Structure-activity relationships for lipoprotein lipase agonists that lower plasma triglycerides in vivo. Eur J Med Chem 2015; 103:191-209. [PMID: 26355531 DOI: 10.1016/j.ejmech.2015.08.058] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 08/25/2015] [Accepted: 08/28/2015] [Indexed: 12/27/2022]
Abstract
The risk of cardiovascular events increases in individuals with elevated plasma triglyceride (TG) levels, therefore advocating the need for efficient TG-lowering drugs. In the blood circulation, TG levels are regulated by lipoprotein lipase (LPL), an unstable enzyme that is only active as a non-covalently associated homodimer. We recently reported on a N-phenylphthalimide derivative (1) that stabilizes LPL in vitro, and moderately lowers triglycerides in vivo (Biochem. Biophys. Res. Commun.2014, 450, 1063). Herein, we establish structure-activity relationships of 51 N-phenylphthalimide analogs of the screening hit 1. In vitro evaluation highlighted that modifications on the phthalimide moiety were not tolerated and that lipophilic substituents on the central phenyl ring were functionally essential. The substitution pattern on the central phenyl ring also proved important to stabilize LPL. However, in vitro testing demonstrated rapid degradation of the phthalimide fragment in plasma which was addressed by replacing the phthalimide scaffold with other heterocyclic fragments. The in vitro potency was retained or improved and substance 80 proved stable in plasma and efficiently lowered plasma TGs in vivo.
Collapse
|
15
|
Cao P, Pan H, Xiao T, Zhou T, Guo J, Su Z. Advances in the Study of the Antiatherogenic Function and Novel Therapies for HDL. Int J Mol Sci 2015. [PMID: 26225968 PMCID: PMC4581191 DOI: 10.3390/ijms160817245] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The hypothesis that raising high-density lipoprotein cholesterol (HDL-C) levels could improve the risk for cardiovascular disease (CVD) is facing challenges. There is multitudinous clear clinical evidence that the latest failures of HDL-C-raising drugs show no clear association with risks for CVD. At the genetic level, recent research indicates that steady-state HDL-C concentrations may provide limited information regarding the potential antiatherogenic functions of HDL. It is evident that the newer strategies may replace therapeutic approaches to simply raise plasma HDL-C levels. There is an urgent need to identify an efficient biomarker that accurately predicts the increased risk of atherosclerosis (AS) in patients and that may be used for exploring newer therapeutic targets. Studies from recent decades show that the composition, structure and function of circulating HDL are closely associated with high cardiovascular risk. A vast amount of data demonstrates that the most important mechanism through which HDL antagonizes AS involves the reverse cholesterol transport (RCT) process. Clinical trials of drugs that specifically target HDL have so far proven disappointing, so it is necessary to carry out review on the HDL therapeutics.
Collapse
Affiliation(s)
- Peiqiu Cao
- Key Research Center of Liver Regulation for Hyperlipemia SATCM/Class III, Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Haitao Pan
- Key Research Center of Liver Regulation for Hyperlipemia SATCM/Class III, Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Tiancun Xiao
- Inorganic Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QR, UK.
- Guangzhou Boxabio Ltd., D-106 Guangzhou International Business Incubator, Guangzhou 510530, China.
| | - Ting Zhou
- Guangzhou Boxabio Ltd., D-106 Guangzhou International Business Incubator, Guangzhou 510530, China.
| | - Jiao Guo
- Key Research Center of Liver Regulation for Hyperlipemia SATCM/Class III, Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhengquan Su
- Key Research Center of Liver Regulation for Hyperlipemia SATCM/Class III, Laboratory of Metabolism SATCM, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
16
|
Garcia GM, Oliveira LT, Pitta IDR, de Lima MDCA, Vilela JMC, Andrade MS, Abdalla DSP, Mosqueira VCF. Improved nonclinical pharmacokinetics and biodistribution of a new PPAR pan-agonist and COX inhibitor in nanocapsule formulation. J Control Release 2015; 209:207-18. [DOI: 10.1016/j.jconrel.2015.04.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 04/16/2015] [Accepted: 04/25/2015] [Indexed: 01/21/2023]
|
17
|
Li Y, He PP, Zhang DW, Zheng XL, Cayabyab FS, Yin WD, Tang CK. Lipoprotein lipase: from gene to atherosclerosis. Atherosclerosis 2014; 237:597-608. [PMID: 25463094 DOI: 10.1016/j.atherosclerosis.2014.10.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 10/13/2014] [Accepted: 10/13/2014] [Indexed: 01/21/2023]
Abstract
Lipoprotein lipase (LPL) is a key enzyme in lipid metabolism and responsible for catalyzing lipolysis of triglycerides in lipoproteins. LPL is produced mainly in adipose tissue, skeletal and heart muscle, as well as in macrophage and other tissues. After synthesized, it is secreted and translocated to the vascular lumen. LPL expression and activity are regulated by a variety of factors, such as transcription factors, interactive proteins and nutritional state through complicated mechanisms. LPL with different distributions may exert distinct functions and have diverse roles in human health and disease with close association with atherosclerosis. It may pose a pro-atherogenic or an anti-atherogenic effect depending on its locations. In this review, we will discuss its gene, protein, synthesis, transportation and biological functions, and then focus on its regulation and relationship with atherosclerosis and potential underlying mechanisms. The goal of this review is to provide basic information and novel insight for further studies and therapeutic targets.
Collapse
Affiliation(s)
- Yuan Li
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Discovery, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China
| | - Ping-Ping He
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Discovery, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China; School of Nursing, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The Cumming School of Medicine, The University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Fracisco S Cayabyab
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Wei-Dong Yin
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Discovery, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Discovery, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
18
|
Bartelt A, Beil FT, Müller B, Koehne T, Yorgan TA, Heine M, Yilmaz T, Rüther W, Heeren J, Schinke T, Niemeier A. Hepatic lipase is expressed by osteoblasts and modulates bone remodeling in obesity. Bone 2014; 62:90-8. [PMID: 24440515 DOI: 10.1016/j.bone.2014.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 12/18/2013] [Accepted: 01/07/2014] [Indexed: 01/23/2023]
Abstract
A number of unexpected molecules were recently identified as products of osteoblasts, linking bone homeostasis to systemic energy metabolism. Here we identify the lipolytic enzyme hepatic lipase (HL, encoded by Lipc) as a novel cell-autonomous regulator of osteoblast function. In an unbiased genome-wide expression analysis, we find Lipc to be highly induced upon osteoblast differentiation, verified by quantitative Taqman analyses of primary osteoblasts in vitro and of bone samples in vivo. Functionally, loss of HL in vitro leads to increased expression and secretion of osteoprotegerin (OPG), while expression of some osteoblast differentiation makers is impaired. When challenging energy metabolism in a diet-induced obesity (DIO) study, lack of HL leads to a significant increase in bone formation markers and a decrease in bone resorption markers. Accordingly, in the DIO setting, we observe in Lipc(-/-) animals but not in wild-type controls a significant increase in lumbar vertebral trabecular bone mass and formation rate as well as in femoral trabecular bone mass and cortical thickness. Taken together, we demonstrate that HL expressed by osteoblasts has an impact on osteoblast OPG expression and that lack of HL leads to increased bone mass in DIO. These data provide a novel and completely unexpected molecular link in the complex interplay of osteoblasts and systemic energy metabolism.
Collapse
Affiliation(s)
- Alexander Bartelt
- Department of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - F Timo Beil
- Department of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Brigitte Müller
- Department of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Till Koehne
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Tayfun Yilmaz
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Wolfgang Rüther
- Department of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Andreas Niemeier
- Department of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| |
Collapse
|
19
|
MicroRNA-27a/b regulates cellular cholesterol efflux, influx and esterification/hydrolysis in THP-1 macrophages. Atherosclerosis 2014; 234:54-64. [PMID: 24608080 DOI: 10.1016/j.atherosclerosis.2014.02.008] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/19/2014] [Accepted: 02/09/2014] [Indexed: 12/20/2022]
Abstract
RATIONALE Macrophage cholesterol homeostasis maintenance is the result of a balance between influx, endogenous synthesis, esterification/hydrolysis and efflux. Excessive accumulation of cholesterol leads to foam cell formation, which is the major pathology of atherosclerosis. Previous studies have shown that miR-27 (miR-27a and miR-27b) may play a key role in the progression of atherosclerosis. OBJECTIVE We set out to investigate the molecular mechanisms of miR-27a/b in intracellular cholesterol homeostasis. METHODS AND RESULTS In the present study, our results have shown that the miR-27 family is highly conserved during evolution, present in mammals and directly targets the 3' UTR of ABCA1, LPL, and ACAT1. apoA1, ABCG1 and SR-B1 lacking miR-27 bind sites should not be influenced by miR-27 directly. miR-27a and miR-27b directly regulated the expression of endogenous ABCA1 in different cells. Treatment with miR-27a and miR-27b mimics reduced apoA1-mediated cholesterol efflux by 33.08% and 44.61% in THP-1 cells, respectively. miR-27a/b also regulated HDL-mediated cholesterol efflux in THP-1 macrophages and affected the expression of apoA1 in HepG2 cells. However, miR-27a/b had no effect on total cellular cholesterol accumulation, but regulated the levels of cellular free cholesterol and cholesterol ester. We further found that miR-27a/b regulated the expression of LPL and CD36, and then affected the ability of THP-1 macrophages to uptake Dil-oxLDL. Finally, we identified that miR-27a/b regulated cholesterol ester formation by targeting ACAT1 in THP-1 macrophages. CONCLUSION These findings indicate that miR-27a/b affects the efflux, influx, esterification and hydrolysis of cellular cholesterol by regulating the expression of ABCA1, apoA1, LPL, CD36 and ACAT1.
Collapse
|
20
|
Bouvy-Liivrand M, Heinäniemi M, John E, Schneider JG, Sauter T, Sinkkonen L. Combinatorial regulation of lipoprotein lipase by microRNAs during mouse adipogenesis. RNA Biol 2014; 11:76-91. [PMID: 24457907 PMCID: PMC3929427 DOI: 10.4161/rna.27655] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 12/20/2013] [Accepted: 12/23/2013] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs (miRNAs) regulate gene expression directly through base pairing to their targets or indirectly through participating in multi-scale regulatory networks. Often miRNAs take part in feed-forward motifs where a miRNA and a transcription factor act on shared targets to achieve accurate regulation of processes such as cell differentiation. Here we show that the expression levels of miR-27a and miR-29a inversely correlate with the mRNA levels of lipoprotein lipase (Lpl), their predicted combinatorial target, and its key transcriptional regulator peroxisome proliferator-activated receptor gamma (Pparg) during 3T3-L1 adipocyte differentiation. More importantly, we show that Lpl, a key lipogenic enzyme, can be negatively regulated by the two miRNA families in a combinatorial fashion on the mRNA and functional level in maturing adipocytes. This regulation is mediated through the Lpl 3'UTR as confirmed by reporter gene assays. In addition, a small mathematical model captures the dynamics of this feed-forward motif and predicts the changes in Lpl mRNA levels upon network perturbations. The obtained results might offer an explanation to the dysregulation of LPL in diabetic conditions and could be extended to quantitative modeling of regulation of other metabolic genes under similar regulatory network motifs.
Collapse
Affiliation(s)
- Maria Bouvy-Liivrand
- Life Sciences Research Unit; University of Luxembourg; Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine; University of Luxembourg; Esch-Sur-Alzette, Luxembourg
| | - Merja Heinäniemi
- Life Sciences Research Unit; University of Luxembourg; Luxembourg, Luxembourg
- Institute of Biomedicine; School of Medicine; University of Eastern Finland; Kuopio, Finland
| | - Elisabeth John
- Life Sciences Research Unit; University of Luxembourg; Luxembourg, Luxembourg
| | - Jochen G Schneider
- Luxembourg Centre for Systems Biomedicine; University of Luxembourg; Esch-Sur-Alzette, Luxembourg
- Saarland University Medical Center; Department of Medicine II; Homburg, Saar, Germany
| | - Thomas Sauter
- Life Sciences Research Unit; University of Luxembourg; Luxembourg, Luxembourg
| | - Lasse Sinkkonen
- Life Sciences Research Unit; University of Luxembourg; Luxembourg, Luxembourg
| |
Collapse
|
21
|
Hagberg C, Mehlem A, Falkevall A, Muhl L, Eriksson U. Endothelial fatty acid transport: role of vascular endothelial growth factor B. Physiology (Bethesda) 2014; 28:125-34. [PMID: 23455771 DOI: 10.1152/physiol.00042.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dietary lipids present in the circulation have to be transported through the vascular endothelium to be utilized by tissue cells, a vital mechanism that is still poorly understood. Vascular endothelial growth factor B (VEGF-B) regulates this process by controlling the expression of endothelial fatty acid transporter proteins (FATPs). Here, we summarize research on the role of the vascular endothelium in nutrient transport, with emphasis on VEGF-B signaling.
Collapse
|
22
|
Therapeutic expression of hairpins targeting apolipoprotein B100 induces phenotypic and transcriptome changes in murine liver. Gene Ther 2013; 21:60-70. [PMID: 24152580 PMCID: PMC3881031 DOI: 10.1038/gt.2013.58] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 08/13/2013] [Accepted: 09/16/2013] [Indexed: 12/23/2022]
Abstract
Constitutive expression of short hairpin RNAs (shRNAs) may cause cellular toxicity in vivo and using microRNA (miRNA) scaffolds can circumvent this problem. Previously, we have shown that embedding small interfering RNA sequences targeting apolipoprotein B100 (ApoB) in shRNA (shApoB) or miRNA (miApoB) scaffolds resulted in differential processing and long-term efficacy in vivo. Here we show that adeno-associated virus (AAV)-shApoB- or AAV-miApoB-mediated ApoB knockdown induced differential liver morphology and transcriptome expression changes. Our analyses indicate that ApoB knockdown with both shApoB and miApoB resulted in alterations of genes involved in lipid metabolism. In addition, in AAV-shApoB-injected animals, genes involved in immune system activation or cell growth and death were affected, which was associated with increased hepatocyte proliferation. Subsequently, in AAV-miApoB-injected animals, changes of genes involved in oxidoreductase activity, oxidative phosphorylation and nucleic bases biosynthetic processes were observed. Our results demonstrate that long-term knockdown of ApoB in vivo by shApoB or miApoB induces several transcriptome changes in murine liver. The increased hepatocyte profileration by AAV-shRNA may have severe long-term effects indicating that AAV-mediated RNA interference therapy using artificial miRNA may be a safer approach for familial hypercholesterolemia therapy.
Collapse
|
23
|
Larsson M, Vorrsjö E, Talmud P, Lookene A, Olivecrona G. Apolipoproteins C-I and C-III inhibit lipoprotein lipase activity by displacement of the enzyme from lipid droplets. J Biol Chem 2013; 288:33997-34008. [PMID: 24121499 DOI: 10.1074/jbc.m113.495366] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Apolipoproteins (apo) C-I and C-III are known to inhibit lipoprotein lipase (LPL) activity, but the molecular mechanisms for this remain obscure. We present evidence that either apoC-I or apoC-III, when bound to triglyceride-rich lipoproteins, prevent binding of LPL to the lipid/water interface. This results in decreased lipolytic activity of the enzyme. Site-directed mutagenesis revealed that hydrophobic amino acid residues centrally located in the apoC-III molecule are critical for attachment to lipid emulsion particles and consequently inhibition of LPL activity. Triglyceride-rich lipoproteins stabilize LPL and protect the enzyme from inactivating factors such as angiopoietin-like protein 4 (angptl4). The addition of either apoC-I or apoC-III to triglyceride-rich particles severely diminished their protective effect on LPL and rendered the enzyme more susceptible to inactivation by angptl4. These observations were seen using chylomicrons as well as the synthetic lipid emulsion Intralipid. In the presence of the LPL activator protein apoC-II, more of apoC-I or apoC-III was needed for displacement of LPL from the lipid/water interface. In conclusion, we show that apoC-I and apoC-III inhibit lipolysis by displacing LPL from lipid emulsion particles. We also propose a role for these apolipoproteins in the irreversible inactivation of LPL by factors such as angptl4.
Collapse
Affiliation(s)
- Mikael Larsson
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, SE-901 87 Umeå, Sweden
| | - Evelina Vorrsjö
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, SE-901 87 Umeå, Sweden
| | - Philippa Talmud
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, 5 University Street, London WC1E 6JF, United Kingdom
| | - Aivar Lookene
- Department of Chemistry, Tallinn University of Technology, Tallinn 12618, Estonia
| | - Gunilla Olivecrona
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, SE-901 87 Umeå, Sweden.
| |
Collapse
|
24
|
Postprandial lipoproteins and the molecular regulation of vascular homeostasis. Prog Lipid Res 2013; 52:446-64. [DOI: 10.1016/j.plipres.2013.06.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 12/17/2022]
|
25
|
Innis SM, Nelson CM. Dietary triacyglycerols rich in sn-2 palmitate alter post-prandial lipoprotein and unesterified fatty acids in term infants. Prostaglandins Leukot Essent Fatty Acids 2013; 89:145-51. [PMID: 23541418 DOI: 10.1016/j.plefa.2013.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human milk TAG contain 20-25% 16:0, with over 70% of the 16:0 at the TAG sn-2 position. The benefits of TAG sn-2 16:0 have been ascribed to reducing 16:0 excretion as insoluble fatty acid soaps. This study builds on knowledge that infants conserve milk TG sn-2 16:0 post-absorption. Comparison of plasma lipids from 120 day old infants fed formula containing 25-27% 16:0 with 29% 16:0 or 5% 16:0 at the TAG sn-2 position showed higher formula sn-2 16:0 led to lower 18:1n-9, but higher 18:2n-6 and 22:6n-3 in the infant plasma unesterified fatty acids, higher 18:0 in LDL TAG, and higher apo B and lower apo A-1. TAG-sn-2 16:0 may provide 16:0 in remnant particles for hepatic elongation to 18:0, needed for plasma and tissue phospholipids. We suggest attention to the plasma unesterified fatty acids as possible sources of fatty acids for membrane phospholipid synthesis.
Collapse
Affiliation(s)
- Sheila M Innis
- Nutrition and Metabolism Research Program, Child and Family Research Institute, Department of Paediatrics, University of British Columbia, Vancouver, BC, Canada V5Z 4H4.
| | | |
Collapse
|
26
|
Overgaard M, Brasen CL, Svaneby D, Feddersen S, Nybo M. Familial lipoprotein lipase deficiency: a case of compound heterozygosity of a novel duplication (R44Kfs*4) and a common mutation (N291S) in the lipoprotein lipase gene. Ann Clin Biochem 2013; 50:374-9. [DOI: 10.1177/0004563213477393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Familial lipoprotein lipase (LPL) deficiency (FLLD) is a rare autosomal recessive genetic disorder caused by homozygous or compound heterozygous mutations in the LPL gene. FLLD individuals usually express an impaired or non-functional LPL enzyme with low or absent triglyceride (TG) hydrolysis activity causing severe hypertriglyceridaemia. Here we report a case of FLLD in a 29-year-old man, who initially presented with eruptive cutaneous xanthomata, elevated plasma TG concentration but no other co-morbidities. Subsequent genetic testing of the patient revealed compound heterozygosity of a novel duplication (p.R44Kfs*4) leading to a premature stop codon in exon 2 and a known mutation (N291S) in exon 5 of the LPL gene. Further biochemical analysis of the patient's postheparin plasma confirmed a reduction of total lipase activity compared with his heterozygous father carrying the common N291S mutation and to a healthy control. Also the patient showed increased (1.85-fold) activity of hepatic lipase (HL), indicating a functional link between HL and LPL. In summary, we report a case of FLLD caused by compound heterozygosity of a new duplication and a common mutation in the LPL gene, resulting in residual LPL activity. With such mutations, individuals may not receive a diagnosis before classical FLLD symptoms appear later in adulthood. Nevertheless, early diagnosis and lipid-lowering treatment may favour a reduced risk of premature cardiovascular disease or acute pancreatitis in such individuals.
Collapse
Affiliation(s)
- Martin Overgaard
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Claus Lohman Brasen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Dea Svaneby
- Department of Clinical Genetics, Vejle Sygehus, Vejle, Denmark
| | - Søren Feddersen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Mads Nybo
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
27
|
Hepatic lipase- and endothelial lipase-deficiency in mice promotes macrophage-to-feces RCT and HDL antioxidant properties. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:691-7. [DOI: 10.1016/j.bbalip.2013.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 12/30/2012] [Accepted: 01/03/2013] [Indexed: 11/22/2022]
|
28
|
Jiang ZG, Robson SC, Yao Z. Lipoprotein metabolism in nonalcoholic fatty liver disease. J Biomed Res 2012; 27:1-13. [PMID: 23554788 PMCID: PMC3596749 DOI: 10.7555/jbr.27.20120077] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/23/2012] [Accepted: 08/29/2012] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), an escalating health problem worldwide, covers a spectrum of pathologies characterized by fatty accumulation in hepatocytes in early stages, with potential progression to liver inflammation, fibrosis, and failure. A close, yet poorly understood link exists between NAFLD and dyslipidemia, a constellation of abnormalities in plasma lipoproteins including triglyceride-rich very low density lipoproteins. Apolipoproteins are a group of primarily liver-derived proteins found in serum lipoproteins; they not only play an extracellular role in lipid transport between vital organs through circulation, but also play an important intracellular role in hepatic lipoprotein assembly and secretion. The liver functions as the central hub for lipoprotein metabolism, as it dictates lipoprotein production and to a significant extent modulates lipoprotein clearance. Lipoprotein metabolism is an integral component of hepatocellular lipid homeostasis and is implicated in the pathogenesis, potential diagnosis, and treatment of NAFLD.
Collapse
Affiliation(s)
- Zhenghui Gordon Jiang
- Department of Medicine, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
29
|
Abstract
Hypertriglyceridemia is a prevalent risk factor for cardiovascular disease (CVD) and increasingly important in the setting of current obesity and insulin resistance epidemics. High triglyceride (TG) levels are markers for several types of atherogenic lipoproteins. Patients who have hypertriglyceridemia may be at significant risk for CVD even if low-density lipoprotein cholesterol levels are at goal, and therefore warrant treatment that optimizes diet, reduces overweight, and promotes regular exercise. High-risk patients with hypertriglyceridemia, such as those with diabetes, CVD, or metabolic syndrome, may benefit from additional drug treatment aside from a statin to address other lipid abnormalities. In this discussion, we review the role of hypertriglyceridemia and its associated atherogenic lipoproteins in the pathogenesis of atherosclerosis, the relevance of a high TG level as a predictor of CVD, the cardiovascular outcomes from TG-lowering intervention trials, and the current guidelines for treating hypertriglyceridemia.
Collapse
|
30
|
Bartelt A, Merkel M, Heeren J. A new, powerful player in lipoprotein metabolism: brown adipose tissue. J Mol Med (Berl) 2012; 90:887-93. [PMID: 22231746 DOI: 10.1007/s00109-012-0858-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 12/14/2011] [Accepted: 12/28/2011] [Indexed: 12/18/2022]
Abstract
Important causes for modern epidemics such as obesity, diabetes, and cardiovascular disease are over- and malnutrition. Dietary as well as endogenous lipids are transported through the bloodstream in lipoproteins, and disturbances in lipoprotein metabolism are associated with atherosclerosis, heart disease, and diabetes. Recent findings reveal biological principles-how lipoproteins, in particular triglyceride-rich lipoproteins, are metabolized and what factors regulate their processing. The fate of triglycerides delivered by lipoproteins is quite simple: either they can be stored or they can be utilized for combustion or biosynthetic pathways. In the healthy state, fatty acids derived from triglycerides can be burned in the heart, muscle, and other organs for actual work load, or they can be stored in white adipose tissue. The combination of storage and combustion is realized in brown adipose tissue (BAT), a peripheral organ that was long thought to be only of relevance in small mammals: Recent data however prove that BAT plays an important role in human adults. Here, we will review recent insights on how BAT controls triglyceride clearance and the possible implications for the treatment of chronic diseases caused by lipid mishandling.
Collapse
Affiliation(s)
- Alexander Bartelt
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | | | | |
Collapse
|
31
|
Protection Against Myocardial Infarction and No-Reflow Through Preservation of Vascular Integrity by Angiopoietin-Like 4. Circulation 2012; 125:140-9. [DOI: 10.1161/circulationaha.111.049072] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background—
Increased permeability, predominantly controlled by endothelial junction stability, is an early event in the deterioration of vascular integrity in ischemic disorders. Hemorrhage, edema, and inflammation are the main features of reperfusion injuries, as observed in acute myocardial infarction (AMI). Thus, preservation of vascular integrity is fundamental in ischemic heart disease. Angiopoietins are pivotal modulators of cell–cell junctions and vascular integrity. We hypothesized that hypoxic induction of angiopoietin-like protein 4 (ANGPTL4) might modulate vascular damage, infarct size, and no-reflow during AMI.
Methods and Results—
We showed that vascular permeability, hemorrhage, edema, inflammation, and infarct severity were increased in
angptl4
-deficient mice. We determined that decrease in vascular endothelial growth factor receptor 2 (VEGFR2) and VE-cadherin expression and increase in Src kinase phosphorylation downstream of VEGFR2 were accentuated after ischemia-reperfusion in the coronary microcirculation of
angptl4
-deficient mice. Both events led to altered VEGFR2/VE-cadherin complexes and to disrupted adherens junctions in the endothelial cells of
angptl4
-deficient mice that correlated with increased no-reflow. In vivo injection of recombinant human ANGPTL4 protected VEGF-driven dissociation of the VEGFR2/VE-cadherin complex, reduced myocardial infarct size, and the extent of no-reflow in mice and rabbits.
Conclusions—
These data showed that ANGPTL4 might constitute a relevant target for therapeutic vasculoprotection aimed at counteracting the effects of VEGF, thus being crucial for preventing no-reflow and conferring secondary cardioprotection during AMI.
Collapse
|
32
|
Lam V, Henault M, Khougaz K, Fortin LJ, Ouellet M, Melnyk R, Partridge A. Resorufin Butyrate as a Soluble and Monomeric High-Throughput Substrate for a Triglyceride Lipase. ACTA ACUST UNITED AC 2011; 17:245-51. [DOI: 10.1177/1087057111422944] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Triglyceride lipases such as lipoprotein lipase, endothelial lipase, and hepatic lipase play key roles in controlling the levels of plasma lipoprotein. Accordingly, small-molecule modulation of these species could alter patient lipid profiles with corresponding health effects. Screening of these enzymes for small-molecule therapeutics has historically involved the use of lipid-based particles to mimic native substrates. However, particle-based artifacts can complicate the discovery of therapeutic molecules. As a simplifying solution, the authors sought to develop an approach involving a soluble and monomeric lipase substrate. Using purified bovine lipoprotein lipase as a model system, they show that the hydrolysis of resorufin butyrate can be fluorescently monitored to give a robust assay (Z′ > 0.8). Critically, using parallel approaches, they show that resorufin butyrate is soluble and monomeric under assay conditions. The presented assay should be useful as a simple and inexpensive primary or secondary screen for the discovery of therapeutic lipase modulators.
Collapse
Affiliation(s)
- Vincent Lam
- Department of In Vitro Sciences, Merck Frosst Centre for Therapeutic Research, Kirkland, Quebec, Canada
| | - Martin Henault
- Department of In Vitro Sciences, Merck Frosst Centre for Therapeutic Research, Kirkland, Quebec, Canada
| | - Karine Khougaz
- Department of Basic Pharmaceutical Sciences, Merck Frosst Centre for Therapeutic Research, Kirkland, Quebec, Canada
| | - Louis-Jacques Fortin
- Department of In Vitro Sciences, Merck Frosst Centre for Therapeutic Research, Kirkland, Quebec, Canada
| | - Marc Ouellet
- Department of In Vitro Sciences, Merck Frosst Centre for Therapeutic Research, Kirkland, Quebec, Canada
| | - Roman Melnyk
- Department of Program Biology, Merck Frosst Centre for Therapeutic Research, Kirkland, Quebec, Canada
| | | |
Collapse
|
33
|
Goldberg IJ, Eckel RH, McPherson R. Triglycerides and heart disease: still a hypothesis? Arterioscler Thromb Vasc Biol 2011; 31:1716-25. [PMID: 21527746 DOI: 10.1161/atvbaha.111.226100] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The purpose of this article is to review the basic and clinical science relating plasma triglycerides and cardiovascular disease. Although many aspects of the basic physiology of triglyceride production, its plasma transport, and its tissue uptake have been known for several decades, the relationship of plasma triglyceride levels to vascular disease is uncertain. Are triglyceride-rich lipoproteins, their influence on high-density lipoprotein and low-density lipoprotein, or the underlying diseases that lead to defects in triglyceride metabolism the culprit? Animal models have failed to confirm that anything other than early fatty lesions can be produced by triglyceride-rich lipoproteins. Metabolic products of triglyceride metabolism can be toxic to arterial cells; however, these studies are primarily in vitro. Correlative studies of fasting and postprandial triglycerides and genetic diseases implicate very-low-density lipoprotein and their remnants and chylomicron remnants in atherosclerosis development, but the concomitant alterations in other lipoproteins and other risk factors obscure any conclusions about direct relationships between disease and triglycerides. Genes that regulate triglyceride levels also correlate with vascular disease. Human intervention trials, however, have lacked an appropriately defined population and have produced outcomes without definitive conclusions. The time is more than ripe for new and creative approaches to understanding the relationship of triglycerides and heart disease.
Collapse
Affiliation(s)
- Ira J Goldberg
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | | | |
Collapse
|
34
|
Basu D, Manjur J, Jin W. Determination of lipoprotein lipase activity using a novel fluorescent lipase assay. J Lipid Res 2011; 52:826-32. [PMID: 21270098 DOI: 10.1194/jlr.d010744] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A novel, real-time, homogeneous fluorogenic lipoprotein lipase (LPL) assay was developed using a commercially available substrate, the EnzChek lipase substrate, which is solubilized in Zwittergent. The triglyceride analog substrate does not fluoresce, owing to apposition of fluorescent and fluorescent quenching groups at the sn-1 and sn-2 positions, respectively, fluorescence becoming unquenched upon release of the sn-1 BODIPY FA derivative following hydrolysis. Increase in fluorescence intensity at 37°C was proportional to LPL concentration. The assay was more sensitive than a similar assay using 1,2-O-dilauryl-rac-glycero-3-glutaric acid-(6-methylresorufin ester) and was validated in biological samples, including determination of LPL-specific activity in postheparin mouse plasma. The simplicity and reproducibility of the assay make it ideal for in vitro, high-throughput screening for inhibitors and activators of LPL, thus expediting discovery of drugs of potential clinical value.
Collapse
Affiliation(s)
- Debapriya Basu
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | | | | |
Collapse
|
35
|
Fuchs B, Süss R, Teuber K, Eibisch M, Schiller J. Lipid analysis by thin-layer chromatography--a review of the current state. J Chromatogr A 2010; 1218:2754-74. [PMID: 21167493 DOI: 10.1016/j.chroma.2010.11.066] [Citation(s) in RCA: 213] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 11/24/2010] [Accepted: 11/26/2010] [Indexed: 12/15/2022]
Abstract
High-performance thin-layer chromatography (HPTLC) is a widely used, fast and relatively inexpensive method of separating complex mixtures. It is particularly useful for smaller, apolar compounds and offers some advantages over HPLC. This review gives an overview about the special features as well as the problems that have to be considered upon the HPTLC analysis of lipids. The term "lipids" is used here in a broad sense and comprises fatty acids and their derivatives as well as substances related biosynthetically or functionally to these compounds. After a short introduction regarding the stationary phases and the methods how lipids can be visualized on an HPTLC plate, the individual lipid classes will be discussed and the most suitable solvent systems for their separation indicated. The focus will be on lipids that are most abundant in biological systems, i.e. cholesterol and its derivates, glycerides, sphingo- and glycolipids as well as phospholipids. Finally, a nowadays very important topic, the combination between HPTLC and mass spectrometric (MS) detection methods will be discussed. It will be shown that this is a very powerful method to investigate the identities of the HPTLC spots in more detail than by the use of common staining methods. Future aspects of HPTLC in the lipid field will be also discussed.
Collapse
Affiliation(s)
- Beate Fuchs
- University of Leipzig, Medical Department, Institute of Medical Physics and Biophysics, Härtelstr. 16/18, D-04107 Leipzig, Germany
| | | | | | | | | |
Collapse
|
36
|
Abstract
Cholesterol-engorged macrophage foam cells are a critical component of the atherosclerotic lesion. Reducing the sterol deposits in lesions reduces clinical events. Sterol accumulations within lysosomes have proven to be particularly hard to mobilize out of foam cells. Moreover, excess sterol accumulation in lysosomes has untoward effects, including a complete disruption of lysosome function. Recently, we demonstrated that treatment of sterol-engorged macrophages in culture with triglyceride-containing particles can reverse many of the effects of cholesterol on lysosomes and dramatically reduce the sterol burden in these cells. This article describes what is known about lysosomal sterol engorgement, discusses the possible mechanisms by which triglyceride could produce its effects, and evaluates the possible positive and negative effects of reducing the lysosomal cholesterol levels in foam cells.
Collapse
Affiliation(s)
- W Gray Jerome
- Department of Pathology, U-2206 Medical Center North Vanderbilt University School of Medicine 1161 21st Avenue, South Nashville, TN 37232-32561, USA, Tel.: +1 615 322 5530
| |
Collapse
|