1
|
Ghasemzadeh Rahbardar M, Fazeli Kakhki H, Hosseinzadeh H. Ziziphus jujuba (Jujube) in Metabolic Syndrome: From Traditional Medicine to Scientific Validation. Curr Nutr Rep 2024; 13:845-866. [PMID: 39354208 DOI: 10.1007/s13668-024-00581-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE OF REVIEW This review evaluates the therapeutic potential of Ziziphus jujuba and its main components in managing complications of metabolic syndrome, including diabetes, dyslipidemia, obesity, and hypertension. RECENT FINDINGS The reviewed studies provide evidence supporting the use of Z. jujuba and its main components (lupeol and betulinic acid) as natural treatments for complications of metabolic syndrome. These substances enhance glucose uptake through the activation of signaling pathways such as phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), reduce hepatic glucose synthesis, and increase glucose uptake by adipocytes and skeletal muscle cells. They also improve insulin sensitivity by modulating AMP-activated protein kinase (AMPK) activity and regulating insulin signaling proteins and glucose transporters. In the field of dyslipidemia, they inhibit triglyceride synthesis, lipid accumulation, and adipogenic enzymes, while influencing key signaling pathways involved in adipogenesis. Z. jujuba and its constituents demonstrate anti-adipogenic effects, inhibiting lipid accumulation and modulating adipogenic enzymes and transcription factors. They also exhibit positive effects on endothelial function and vascular health by enhancing endothelial nitric oxide synthase (eNOS) expression, NO production, and antioxidant enzyme activity. Z. jujuba, lupeol, and betulinic acid hold promise as natural treatments for complications of metabolic syndrome. They improve glucose metabolism, insulin sensitivity, and lipid profiles while exerting anti-adipogenic effects and enhancing endothelial function. However, further research is needed to elucidate the mechanisms and confirm their efficacy in clinical trials. These natural compounds offer potential as alternative therapies for metabolic disorders and contribute to the growing body of evidence supporting the use of natural medicines in their management.
Collapse
Affiliation(s)
| | - Homa Fazeli Kakhki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Davidson WS, Vaisar T, Heinecke JW, Bornfeldt KE. Distinct roles of size-defined HDL subpopulations in cardiovascular disease. Curr Opin Lipidol 2024:00041433-990000000-00093. [PMID: 39450930 DOI: 10.1097/mol.0000000000000959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW Doubts about whether high-density lipoprotein-cholesterol (HDL-C) levels are causally related to atherosclerotic cardiovascular disease (CVD) risk have stimulated research on identifying HDL-related metrics that might better reflect its cardioprotective functions. HDL is made up of different types of particles that vary in size, protein and lipid composition, and function. This review focuses on recent findings on the specific roles of HDL subpopulations defined by size in CVD. RECENT FINDINGS Small HDL particles are more effective than larger particles at promoting cellular cholesterol efflux because apolipoprotein A-I on their surface better engages ABCA1 (ATP binding cassette subfamily A member 1). In contrast, large HDL particles bind more effectively to scavenger receptor class B type 1 on endothelial cells, which helps prevent LDL from moving into the artery wall. The specific role of medium-sized HDL particles, the most abundant subpopulation, is still unclear. SUMMARY HDL is made up of subpopulations of different sizes of particles, with selective functional roles for small and large HDLs. The function of HDL may depend more on the size and composition of its subpopulations than on HDL-C levels. Further research is required to understand how these different HDL subpopulations influence the development of CVD.
Collapse
Affiliation(s)
- W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Tomas Vaisar
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
| | - Jay W Heinecke
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
| | - Karin E Bornfeldt
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
3
|
Wu W, Jian Y, Yuan S, Li X, Tang Y, Zeng F, Liu W, Zhao Z, Wang Y, Wang Y, Liu W. Exercise-promoted adiponectin secretion activates autolysosomes to protect the liver of ApoE -/- mice from a high-fat diet. Food Funct 2024; 15:9796-9812. [PMID: 39229645 DOI: 10.1039/d4fo02984d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Fat is a "double-edged sword": while it is a necessary substance for the body, the long-term intake of excessive fat will cause obesity, with the liver subjected to lipotoxicity as it accumulates. It will then continue to deteriorate, eventually leading to liver failure, which is a negative impact of high-fat food intake. Research has shown that exercise can reverse the side effects of a chronic high-fat diet and help the body to mitigate the harmful effects of lipotoxicity. In our study, it was found that moderate-intensity cardio-training (MICT) and high-intensity interval exercise (HIIT) effectively protected the livers of high-fat diet (HFD) ApoE-/- mice against lipotoxicity. Previous results demonstrated that 12 weeks of HFD resulted in a significant elevation of CD36 in the livers of C57BL/6J mice, while knockdown of CD36 did not reduce the accumulation of fat in the liver. Therefore, we used ApoE-/- mice as experimental subjects. Although HFD caused the development of hyperlipidemia and atherosclerosis, it is interesting to note that, due to the knockdown of ApoE, the livers of ApoE-/- mice in the non-exercise group did not show significant lipid deposition; however, after 12 weeks of MICT and HIIT, the livers of ApoE-/- mice showed significant lipid deposition. After we analyzed the lipid metabolism in their livers, we found that this was caused by the promotion of transport of peripheral fat into the liver due to exercise. Moreover, 12 weeks of exercise effectively reduced atherosclerosis, and the livers of ApoE-/- mice in the exercise group were not damaged by lipotoxicity. The results showed that a 12-week exercise treatment activated AMPK in the livers of HFD ApoE-/- mice through the APN-AdipoR1 signaling pathway, improved hepatic lipid metabolism disorders, and promoted the nuclear translocation of TFEB to enhance autophagic-lysosomal lipid scavenging. After the peripheral lipid is input into the liver due to exercise, the energy generated through gluconeogenesis can be used to replenish the energy consumed by exercise and maintain the normal operation of various functions in the liver, based on which the high autophagic flux in the liver can be maintained and the lipid clearance rate can be enhanced to protect the liver from lipotoxicity.
Collapse
Affiliation(s)
- Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Ye Jian
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Shunling Yuan
- Yangtze University, College of Arts and Sciences, Jingzhou 434020, China
| | - Xuan Li
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yingzhe Tang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Wenjing Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zhe Zhao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yirong Wang
- Hunan Sports Vocational College, Changsha 410019, China
| | - Yiyang Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| |
Collapse
|
4
|
Khalil A. Nutrition, Lipoproteins and Cardiovascular Diseases. Nutrients 2024; 16:2530. [PMID: 39125409 PMCID: PMC11314000 DOI: 10.3390/nu16152530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 08/12/2024] Open
Abstract
Cardiovascular diseases (CVDs) represent the leading cause of mortality worldwide, despite the significant advancements that have been made in terms of primary and secondary prevention strategies over the past decades [...].
Collapse
Affiliation(s)
- Abdelouahed Khalil
- Geriatrics Unit, Department of Medicine, Faculty of Medicine and Biological Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| |
Collapse
|
5
|
Xu C, Meng J, Yu XH, Wang RJ, Li ML, Yin SH, Wang G. TNFAIP1 promotes macrophage lipid accumulation and accelerates the development of atherosclerosis through the LEENE/FoxO1/ABCA1 pathway. J Physiol Biochem 2024; 80:523-539. [PMID: 38878215 DOI: 10.1007/s13105-024-01018-x] [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: 09/13/2023] [Accepted: 03/12/2024] [Indexed: 10/25/2024]
Abstract
Macrophage lipid accumulation is a critical contributor to foam cell formation and atherosclerosis. Tumor necrosis factor-α-induced protein 1 (TNFAIP1) is closely associated with cardiovascular disease. However, its role and molecular mechanisms in atherogenesis remain unclear. TNFAIP1 was knocked down in THP-1 macrophage-derived foam cells and apolipoprotein-deficient (apoE-/-) mice using lentiviral vector. The expression of lncRNA enhancing endothelial nitric oxide synthase expression (LEENE), Forkhead box O1 (FoxO1) and ATP binding cassette transporter A1 (ABCA1) was evaluated by qRT-PCR and/or western blot. Lipid accumulation in macrophage was assessed by high-performance liquid chromatography and Oil red O staining. RNA immunoprecipitation and RNA pull-down assay were performed to verify the interaction between LEENE and FoxO1 protein. Atherosclerotic lesions were analyzed using HE, Oil red O and Masson staining. Our results showed that TNFAIP1 was significantly increased in THP-1 macrophages loaded with oxidized low-density lipoprotein. Knockdown of TNFAIP1 enhanced LEENE expression, promoted the direct interaction of LEENE with FoxO1 protein, stimulated FoxO1 protein degradation through the proteasome pathway, induced ABCA1 transcription, and finally suppressed lipid accumulation in THP-1 macrophage-derived foam cells. TNFAIP1 knockdown also up-regulated ABCA1 expression, improved plasma lipid profiles, enhanced the efficiency of reverse cholesterol transport and attenuated lesion area in apoE-/- mice. Taken together, these results provide the first direct evidence that TNFAIP1 aggravates atherosclerosis by promoting macrophage lipid accumulation via the LEENE/FoxO1/ABCA1 signaling pathway. TNFAIP1 may represent a promising therapeutic target for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Can Xu
- Department of Cardiology, Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Jun Meng
- Department of Function, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, 570100, Hainan, People's Republic of China
| | - Ru-Jing Wang
- Department of Cardiology, Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Mei-Ling Li
- Department of Cardiology, Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Shan-Hui Yin
- Department of Neonatology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| | - Gang Wang
- Department of Cardiology, Institute of Cardiovascular Disease, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
6
|
Harsch BA, Borkowski K, Walker RE, Pedersen TL, Newman JW, Shearer GC. ABCA1 and apoA-I dependent 12-hydroxyeicosatetraenoic acid efflux regulates macrophage inflammatory signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603001. [PMID: 39026807 PMCID: PMC11257534 DOI: 10.1101/2024.07.11.603001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Aberrant high-density lipoprotein (HDL) function is implicated in inflammation-associated pathologies. While HDL ABCA1-mediated reverse cholesterol and phospholipid transport are well described, the movement of pro-/anti-inflammatory lipids has not been explored. HDL phospholipids are the largest reservoir of circulating arachidonic acid-derived oxylipins. Endotoxin-stimulation activates inflammatory cells leading to hydroxyeicosatetraenoic acid (HETE) production, oxylipins which are involved in inflammatory response coordination. Active signaling in the non-esterified (NE) pool is terminated by sequestration of HETEs as esterified (Es) forms and degradation. We speculate that an ABCA1-apoA-I-dependent efflux of HETEs from stimulated cells could regulate intracellular HETE availability. Here we test this hypothesis both in vitro and in vivo. In endotoxin-stimulated RAW-264.7 macrophages preloaded with d8-arachidonic acid we use compartmental tracer modeling to characterize the formation of HETEs, and their efflux into HDL. We found that in response to endotoxin: I) Cellular NE 12-HETE is positively associated with MCP-1 secretion (p<0.001); II) HETE transfer from NE to Es pools is ABCA1-depedent (p<0.001); III) Cellular Es HETEs are transported into media when both apoA-I and ABCA1 are present (p<0.001); IV) The stimulated efflux of HETEs >> arachidonate (p<0.001). Finally, in endotoxin challenged humans (n=17), we demonstrate that intravenous lipopolysaccharide (0.6 ng/kg body weight) resulted in accumulation of 12-HETE in HDL over a 168-hour follow-up. Therefore, HDL can suppress inflammatory responses in macrophages by regulating intracellular HETE content in an apoA-I/ABCA1 dependent manner. The described mechanism may apply to other oxylipins and explain anti-inflammatory properties of HDL. This newly defined HDL property opens new doors for the study of lipoprotein interactions in metabolic diseases.
Collapse
Affiliation(s)
- Brian A Harsch
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - Kamil Borkowski
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis CA
| | - Rachel E Walker
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | | | - John W Newman
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis CA
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis CA
- Department of Nutrition, University of California Davis, Davis CA
| | - Gregory C Shearer
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| |
Collapse
|
7
|
Povsic TJ, Korjian S, Bahit MC, Chi G, Duffy D, Alexander JH, Vinereanu D, Tricoci P, Mears SJ, Deckelbaum LI, Bonaca M, Ridker PM, Goodman SG, Cornel JH, Lewis BS, Parkhomenko A, Lopes RD, Aylward P, Lincoff AM, Heise M, Sacks F, Nicolau JC, Merkely B, Trebacz J, Libby P, Nicholls SJ, Pocock S, Bhatt DL, Kastelein J, Bode C, Mahaffey KW, Steg PG, Tendera M, Bainey KR, Harrington RA, Mehran R, Duerschmied D, Kingwell BA, Gibson CM. Effect of Reconstituted Human Apolipoprotein A-I on Recurrent Ischemic Events in Survivors of Acute MI. J Am Coll Cardiol 2024; 83:2163-2174. [PMID: 38588930 DOI: 10.1016/j.jacc.2024.03.396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND The AEGIS-II trial hypothesized that CSL112, an intravenous formulation of human apoA-I, would lower the risk of plaque disruption, decreasing the risk of recurrent events such as myocardial infarction (MI) among high-risk patients with MI. OBJECTIVES This exploratory analysis evaluates the effect of CSL112 therapy on the incidence of cardiovascular (CV) death and recurrent MI. METHODS The AEGIS-II trial was an international, multicenter, randomized, double-blind, placebo-controlled trial that randomized 18,219 high-risk acute MI patients to 4 weekly infusions of apoA-I (6 g CSL112) or placebo. RESULTS The incidence of the composite of CV death and type 1 MI was 11% to 16% lower in the CSL112 group over the study period (HR: 0.84; 95% CI: 0.7-1.0; P = 0.056 at day 90; HR: 0.86; 95% CI: 0.74-0.99; P = 0.048 at day 180; and HR: 0.89; 95% CI: 0.79-1.01; P = 0.07 at day 365). Similarly, the incidence of CV death or any MI was numerically lower in CSL112-treated patients throughout the follow-up period (HR: 0.92; 95% CI: 0.80-1.05 at day 90, HR: 0.89; 95% CI: 0.79-0.996 at day 180, HR: 0.91; 95% CI: 0.83-1.01 at day 365). The effect of CSL112 treatment on MI was predominantly observed for type 1 MI and type 4b (MI due to stent thrombosis). CONCLUSIONS Although CSL112 did not significantly reduce the occurrence of the primary study endpoints, patients treated with CSL112 infusions had numerically lower rates of CV death and MI, type-1 MI, and stent thrombosis-related MI compared with placebo. These findings could suggest a role of apoA-I in reducing subsequent plaque disruption events via enhanced cholesterol efflux. Further prospective data would be needed to confirm these observations.
Collapse
Affiliation(s)
- Thomas J Povsic
- Duke Clinical Research Institute/Duke University Medical Center, Durham, North Carolina, USA
| | - Serge Korjian
- PERFUSE Study Group, Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Gerald Chi
- PERFUSE Study Group, Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - John H Alexander
- Duke Clinical Research Institute/Duke University Medical Center, Durham, North Carolina, USA
| | - Dragos Vinereanu
- University of Medicine and Pharmacy Carol Davila, University and Emergency Hospital, Bucharest, Romania
| | | | | | | | - Marc Bonaca
- University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Paul M Ridker
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Jan H Cornel
- Radboud University Medical Center, Nijmegen and Noordwest Ziekenhuisgroep, Alkmaar, the Netherlands
| | - Basil S Lewis
- Lady Davis Carmel Medical Center and the Technion-Israel Institute of Technology, Aurora, Colorado, USA
| | | | - Renato D Lopes
- Duke Clinical Research Institute/Duke University Medical Center, Durham, North Carolina, USA
| | - Philip Aylward
- South Australian Health and Medical Research Institute/SAHMRI, Adelaide, South Australia, Australia
| | - A Michael Lincoff
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland, Ohio, USA
| | - Mark Heise
- CSL Behring, King of Prussia, Pennsylvania, USA
| | - Frank Sacks
- Department of Nutrition, Harvard School of Public Health, Harvard Medical School, Boston, Massachusetts, USA
| | - Jose C Nicolau
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Bela Merkely
- Heart and Vascular Center of Semmelweis University, Budapest, Hungary
| | - Jaroslaw Trebacz
- Krakowski Szpital Specjalistyczny im. Jana Pawła II, Kraków, Poland
| | - Peter Libby
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen J Nicholls
- Victorian Heart Hospital, Monash Heart and Intensive Care, Clayton, Victoria, Australia
| | - Stuart Pocock
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John Kastelein
- Academic Medical Centre/University of Amsterdam, Amsterdam, the Netherlands
| | | | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Stanford University School of Medicine, Stanford, California, USA
| | - P Gabriel Steg
- Universite Paris-Cité, INSERM 1148, FACT, and AP-HP, Hôpital Bichat, Paris, France
| | - Michal Tendera
- Department of Cardiology and Structural Heart Disease, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Kevin R Bainey
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Daniel Duerschmied
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | | | - C Michael Gibson
- PERFUSE Study Group, Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
8
|
Jiang Y, Pang S, Liu X, Wang L, Liu Y. The Gut Microbiome Affects Atherosclerosis by Regulating Reverse Cholesterol Transport. J Cardiovasc Transl Res 2024; 17:624-637. [PMID: 38231373 DOI: 10.1007/s12265-024-10480-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/07/2024] [Indexed: 01/18/2024]
Abstract
The human system's secret organ, the gut microbiome, has received considerable attention. Emerging research has yielded substantial scientific evidence indicating that changes in gut microbial composition and microbial metabolites may contribute to the development of atherosclerotic cardiovascular disease. The burden of cardiovascular disease on healthcare systems is exacerbated by atherosclerotic cardiovascular disease, which continues to be the leading cause of mortality globally. Reverse cholesterol transport is a powerful protective mechanism that effectively prevents excessive accumulation of cholesterol for atherosclerotic cardiovascular disease. It has been revealed how the gut microbiota modulates reverse cholesterol transport in patients with atherosclerotic risk. In this review, we highlight the complex interactions between microbes, their metabolites, and their potential impacts in reverse cholesterol transport. We also explore the feasibility of modulating gut microbes and metabolites to facilitate reverse cholesterol transport as a novel therapy for atherosclerosis.
Collapse
Affiliation(s)
- Yangyang Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuchao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Xiaoyu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lixin Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| |
Collapse
|
9
|
Nagaoka M, Sakai Y, Nakajima M, Fukami T. Role of carboxylesterase and arylacetamide deacetylase in drug metabolism, physiology, and pathology. Biochem Pharmacol 2024; 223:116128. [PMID: 38492781 DOI: 10.1016/j.bcp.2024.116128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/20/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Carboxylesterases (CES1 and CES2) and arylacetamide deacetylase (AADAC), which are expressed primarily in the liver and/or gastrointestinal tract, hydrolyze drugs containing ester and amide bonds in their chemical structure. These enzymes often catalyze the conversion of prodrugs, including the COVID-19 drugs remdesivir and molnupiravir, to their pharmacologically active forms. Information on the substrate specificity and inhibitory properties of these enzymes, which would be useful for drug development and toxicity avoidance, has accumulated. Recently,in vitroandin vivostudies have shown that these enzymes are involved not only in drug hydrolysis but also in lipid metabolism. CES1 and CES2 are capable of hydrolyzing triacylglycerol, and the deletion of their orthologous genes in mice has been associated with impaired lipid metabolism and hepatic steatosis. Adeno-associated virus-mediated human CES overexpression decreases hepatic triacylglycerol levels and increases fatty acid oxidation in mice. It has also been shown that overexpression of CES enzymes or AADAC in cultured cells suppresses the intracellular accumulation of triacylglycerol. Recent reports indicate that AADAC can be up- or downregulated in tumors of various organs, and its varied expression is associated with poor prognosis in patients with cancer. Thus, CES and AADAC not only determine drug efficacy and toxicity but are also involved in pathophysiology. This review summarizes recent findings on the roles of CES and AADAC in drug metabolism, physiology, and pathology.
Collapse
Affiliation(s)
- Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
10
|
Christ C, Ocskay Z, Kovács G, Jakus Z. Characterization of Atherosclerotic Mice Reveals a Sex-Dependent Susceptibility to Plaque Calcification but No Major Changes in the Lymphatics in the Arterial Wall. Int J Mol Sci 2024; 25:4046. [PMID: 38612867 PMCID: PMC11012298 DOI: 10.3390/ijms25074046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Lymphatics participate in reverse cholesterol transport, and their presence in the arterial wall of the great vessels and prior experimental results suggest their possible role in the development of atherosclerosis. The aim of this study was to characterize the lymphatic vasculature of the arterial wall in atherosclerosis. Tissue sections and tissue-cleared aortas of wild-type mice unveiled significant differences in the density of the arterial lymphatic network throughout the arterial tree. Male and female Ldlr-/- and ApoE-/- mice on a Western diet showed sex-dependent differences in plaque formation and calcification. Female mice on a Western diet developed more calcification of atherosclerotic plaques than males. The lymphatic vessels within the aortic wall of these mice showed no major changes regarding the number of lymphatic junctions and end points or the lymphatic area. However, female mice on a Western diet showed moderate dilation of lymphatic vessels in the abdominal aorta and exhibited indications of increased peripheral lymphatic function, findings that require further studies to understand the role of lymphatics in the arterial wall during the development of atherosclerosis.
Collapse
Affiliation(s)
| | | | | | - Zoltán Jakus
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary; (C.C.); (Z.O.); (G.K.)
| |
Collapse
|
11
|
Apalowo OE, Adegoye GA, Mbogori T, Kandiah J, Obuotor TM. Nutritional Characteristics, Health Impact, and Applications of Kefir. Foods 2024; 13:1026. [PMID: 38611332 PMCID: PMC11011999 DOI: 10.3390/foods13071026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
A global epidemiological shift has been observed in recent decades, characterized by an increase in age-related disorders, notably non-communicable chronic diseases, such as type 2 diabetes mellitus, cardiovascular and neurodegenerative diseases, and cancer. An appreciable causal link between changes in the gut microbiota and the onset of these maladies has been recognized, offering an avenue for effective management. Kefir, a probiotic-enriched fermented food, has gained significance in this setting due to its promising resource for the development of functional or value-added food formulations and its ability to reshape gut microbial composition. This has led to increasing commercial interest worldwide as it presents a natural beverage replete with health-promoting microbes and several bioactive compounds. Given the substantial role of the gut microbiota in human health and the etiology of several diseases, we conducted a comprehensive synthesis covering a total of 33 investigations involving experimental animal models, aimed to elucidate the regulatory influence of bioactive compounds present in kefir on gut microbiota and their potential in promoting optimal health. This review underscores the outstanding nutritional properties of kefir as a central repository of bioactive compounds encompassing micronutrients and amino acids and delineates their regulatory effects at deficient, adequate, and supra-nutritional intakes on the gut microbiota and their broader physiological consequences. Furthermore, an investigation of putative mechanisms that govern the regulatory effects of kefir on the gut microbiota and its connections with various human diseases was discussed, along with potential applications in the food industry.
Collapse
Affiliation(s)
- Oladayo Emmanuel Apalowo
- Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Starkville, MS 39762, USA; (O.E.A.); (G.A.A.)
| | - Grace Adeola Adegoye
- Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Starkville, MS 39762, USA; (O.E.A.); (G.A.A.)
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, USA;
| | - Teresia Mbogori
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, USA;
| | - Jayanthi Kandiah
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, USA;
| | | |
Collapse
|
12
|
He Y, Pavanello C, Hutchins PM, Tang C, Pourmousa M, Vaisar T, Song HD, Pastor RW, Remaley AT, Goldberg IJ, Costacou T, Sean Davidson W, Bornfeldt KE, Calabresi L, Segrest JP, Heinecke JW. Flipped C-Terminal Ends of APOA1 Promote ABCA1-Dependent Cholesterol Efflux by Small HDLs. Circulation 2024; 149:774-787. [PMID: 38018436 PMCID: PMC10913861 DOI: 10.1161/circulationaha.123.065959] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/05/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Cholesterol efflux capacity (CEC) predicts cardiovascular disease independently of high-density lipoprotein (HDL) cholesterol levels. Isolated small HDL particles are potent promoters of macrophage CEC by the ABCA1 (ATP-binding cassette transporter A1) pathway, but the underlying mechanisms are unclear. METHODS We used model system studies of reconstituted HDL and plasma from control and lecithin-cholesterol acyltransferase (LCAT)-deficient subjects to investigate the relationships among the sizes of HDL particles, the structure of APOA1 (apolipoprotein A1) in the different particles, and the CECs of plasma and isolated HDLs. RESULTS We quantified macrophage and ABCA1 CEC of 4 distinct sizes of reconstituted HDL. CEC increased as particle size decreased. Tandem mass spectrometric analysis of chemically cross-linked peptides and molecular dynamics simulations of APOA1, the major protein of HDL, indicated that the mobility of C-terminus of that protein was markedly higher and flipped off the surface in the smallest particles. To explore the physiological relevance of the model system studies, we isolated HDL from LCAT-deficient subjects, whose small HDLs (like reconstituted HDLs) are discoidal and composed of APOA1, cholesterol, and phospholipid. Despite their very low plasma levels of HDL particles, these subjects had normal CEC. In both the LCAT-deficient subjects and control subjects, the CEC of isolated extra-small HDL (a mixture of extra-small and small HDL by calibrated ion mobility analysis) was 3- to 5-fold greater than that of the larger sizes of isolated HDL. Incubating LCAT-deficient plasma and control plasma with human LCAT converted extra-small and small HDL particles into larger particles, and it markedly inhibited CEC. CONCLUSIONS We present a mechanism for the enhanced CEC of small HDLs. In smaller particles, the C-termini of the 2 antiparallel molecules of APOA1 are "flipped" off the lipid surface of HDL. This extended conformation allows them to engage with ABCA1. In contrast, the C-termini of larger HDLs are unable to interact productively with ABCA1 because they form a helical bundle that strongly adheres to the lipid on the particle. Enhanced CEC, as seen with the smaller particles, predicts decreased cardiovascular disease risk. Thus, extra-small and small HDLs may be key mediators and indicators of the cardioprotective effects of HDL.
Collapse
Affiliation(s)
- Yi He
- Department of Medicine, University of Washington, Seattle (Y.H., P.M.H., C.T., T.V., K.E.B., J.W.H.)
| | - Chiara Pavanello
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Italy (C.P., L.C.)
| | - Patrick M. Hutchins
- Department of Medicine, University of Washington, Seattle (Y.H., P.M.H., C.T., T.V., K.E.B., J.W.H.)
| | - Chongren Tang
- Department of Medicine, University of Washington, Seattle (Y.H., P.M.H., C.T., T.V., K.E.B., J.W.H.)
| | - Mohsen Pourmousa
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute (M.P., R.W.P.), National Institutes of Health, Bethesda, MD
| | - Tomas Vaisar
- Department of Medicine, University of Washington, Seattle (Y.H., P.M.H., C.T., T.V., K.E.B., J.W.H.)
| | - Hyun D. Song
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (H.D.S., J.P.S.)
| | - Richard W. Pastor
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute (M.P., R.W.P.), National Institutes of Health, Bethesda, MD
| | - Alan T. Remaley
- Department of Laboratory Medicine (A.T.R.), National Institutes of Health, Bethesda, MD
| | - Ira J. Goldberg
- Department of Medicine, New York University, New York, NY (I.J.G.)
| | - Tina Costacou
- Department of Epidemiology, University of Pittsburgh, PA (T.C.)
| | - W. Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, OH (W.S.D.)
| | - Karin E. Bornfeldt
- Department of Medicine, University of Washington, Seattle (Y.H., P.M.H., C.T., T.V., K.E.B., J.W.H.)
| | - Laura Calabresi
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Italy (C.P., L.C.)
| | - Jere P. Segrest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (H.D.S., J.P.S.)
| | - Jay W. Heinecke
- Department of Medicine, University of Washington, Seattle (Y.H., P.M.H., C.T., T.V., K.E.B., J.W.H.)
| |
Collapse
|
13
|
Lai SJ, Kameda T, Morita M, Yamagata Y, Nishizaka K, Horiuchi Y, Kobayashi Y, Usami Y, Liu JJ, Kasama T, Tozuka M, Ohkawa R. Characterization of novel truncated apolipoprotein A-I in human high-density lipoprotein generated by sequential treatment with myeloperoxidase and chymase. Biochimie 2024; 218:34-45. [PMID: 37774825 DOI: 10.1016/j.biochi.2023.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023]
Abstract
High-density lipoprotein (HDL) cholesterol is a well-known biomarker, which has been associated with reduction in the risk of cardiovascular diseases (CVD). However, some HDL anti-atherosclerotic functions may be impaired without altered HDL-cholesterol (HDL-C) level via its dysfunctional proteins or other physiological reactions in vivo. We previously showed that activated mast cell-derived chymase could modestly cleave apolipoprotein A-I (apoA-I) in HDL3, and further easily cleave lipid-free apoA-I. In contrast, myeloperoxidase (MPO) secreted by macrophages, the main cell type in atherosclerotic plaques, could oxidize HDL proteins, which might modify their tertiary structures, increasing their susceptibility to other enzymes. Here we focused on the co-modification and impact of chymase and MPO, usually secreted during inflammation from cells with possible co-existence in atheromas, on HDL. Only after sequential treatment with MPO and then chymase, two novel truncated apoA-I fragments were generated from HDL. One fragment was 16.5 kDa, and the cleavage site by chymase after MPO modification was the C-terminal of Tyr100 in apoA-I, cross-validated by three different mass spectrometry methods. This novel apoA-I fragment can be trapped in HDL particles to avoid kidney glomerular filtration and has a specific site for antibody generation for ELISA tests. As such, its quantification can be useful in predicting patients with CVD having normal HDL-C levels.
Collapse
Affiliation(s)
- Shao-Jui Lai
- Clinical Bioanalysis and Molecular Biology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Takahiro Kameda
- Clinical Bioanalysis and Molecular Biology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Maasa Morita
- Clinical Bioanalysis and Molecular Biology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan; Clinical Laboratory, Keio University Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuka Yamagata
- Clinical Bioanalysis and Molecular Biology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Kaoruko Nishizaka
- Clinical Bioanalysis and Molecular Biology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Yuna Horiuchi
- Clinical Bioanalysis and Molecular Biology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan; Department of Clinical Laboratory Technology, Faculty of Medical Sciences, Juntendo University, 6-8-1, Hinode, Urayasu, Chiba, 279-0013, Japan
| | - Yukihiro Kobayashi
- Department of Laboratory Medicine, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto City, Nagano, 390-8621, Japan
| | - Yoko Usami
- Department of Laboratory Medicine, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto City, Nagano, 390-8621, Japan
| | - Jun-Jen Liu
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, No. 250, Wuxing St., Xinyi Dist., Taipei City, 110301, Taiwan, ROC
| | - Takeshi Kasama
- HiPep Laboratories, 486-46 Nakatsukasa-cho, Kamigyo-ku, Kyoto, 602-8158, Japan
| | - Minoru Tozuka
- Clinical Bioanalysis and Molecular Biology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan; Life Science Research Center, Nagano Children's Hospital, 3100 Toyoshina, Azumino, 399-8288, Japan
| | - Ryunosuke Ohkawa
- Clinical Bioanalysis and Molecular Biology, Field of Applied Laboratory Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
14
|
Steinbauer S, König A, Neuhauser C, Schwarzinger B, Stangl H, Iken M, Weghuber J, Röhrl C. Elder (Sambucus nigra), identified by high-content screening, counteracts foam cell formation without promoting hepatic lipogenesis. Sci Rep 2024; 14:3547. [PMID: 38347122 PMCID: PMC10861454 DOI: 10.1038/s41598-024-54108-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 02/08/2024] [Indexed: 02/15/2024] Open
Abstract
Cholesterol deposition in intimal macrophages leads to foam cell formation and atherosclerosis. Reverse cholesterol transport (RCT), initiated by efflux of excess cholesterol from foam cells, counteracts atherosclerosis. However, targeting RCT by enhancing cholesterol efflux was so far accompanied by adverse hepatic lipogenesis. Here, we aimed to identify novel natural enhancers of macrophage cholesterol efflux suitable for the prevention of atherosclerosis. Plant extracts of an open-access library were screened for their capacity to increase cholesterol efflux in RAW264.7 macrophages trace-labeled with fluorescent BODIPY-cholesterol. Incremental functional validation of hits yielded two final extracts, elder (Sambucus nigra) and bitter orange (Citrus aurantium L.) that induced ATP binding cassette transporter A1 (ABCA1) expression and reduced cholesteryl ester accumulation in aggregated LDL-induced foam cells. Aqueous elder extracts were subsequently prepared in-house and both, flower and leaf extracts increased ABCA1 mRNA and protein expression in human THP-1 macrophages, while lipogenic gene expression in hepatocyte-derived cells was not induced. Chlorogenic acid isomers and the quercetin glycoside rutin were identified as the main polyphenols in elder extracts with putative biological action. In summary, elder flower and leaf extracts increase macrophage ABCA1 expression and reduce foam cell formation without adversely affecting hepatic lipogenesis.
Collapse
Affiliation(s)
- Stefanie Steinbauer
- University of Applied Sciences Upper Austria, Stelzhamerstrasse 23, 4600, Wels, Austria
| | - Alice König
- University of Applied Sciences Upper Austria, Stelzhamerstrasse 23, 4600, Wels, Austria
- Austrian Competence Center for Feed and Food Quality, Safety and Innovation, Wels, Austria
| | - Cathrina Neuhauser
- Austrian Competence Center for Feed and Food Quality, Safety and Innovation, Wels, Austria
| | - Bettina Schwarzinger
- University of Applied Sciences Upper Austria, Stelzhamerstrasse 23, 4600, Wels, Austria
- Austrian Competence Center for Feed and Food Quality, Safety and Innovation, Wels, Austria
| | - Herbert Stangl
- Center for Pathobiochemistry and Genetics, Institute of Medical Chemistry, Medical University of Vienna, Vienna, Austria
| | | | - Julian Weghuber
- University of Applied Sciences Upper Austria, Stelzhamerstrasse 23, 4600, Wels, Austria.
- Austrian Competence Center for Feed and Food Quality, Safety and Innovation, Wels, Austria.
| | - Clemens Röhrl
- University of Applied Sciences Upper Austria, Stelzhamerstrasse 23, 4600, Wels, Austria.
| |
Collapse
|
15
|
Deng WY, Zhou CL, Zeng MY. Gypenoside XVII inhibits ox-LDL-induced macrophage inflammatory responses and promotes cholesterol efflux through activating the miR-182-5p/HDAC9 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117070. [PMID: 37625608 DOI: 10.1016/j.jep.2023.117070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/11/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The deposition of lipids in macrophages and the subsequent formation of foam cells significantly increase the risk of developing atherosclerosis (As). Targeting ATP-binding cassette transporter A1/G1 (ABCA1/ABCG1)-mediated reverse cholesterol transport is crucial for regulating foam cell formation. Therefore, the search for natural chemical components with the ability to regulate ABCA1/G1 is a potential drug target to combat the development of atherosclerosis. Gypenoside XVII (GP-17), a gypenoside monomer extracted from gynostemma pentaphyllum, presents an efficient anti-atherosclerosis function. However, the suppressed formation mechanism of foam cells by GP-17 remains elusive. AIM OF STUDY To explore the protective activities of GP-17 in ox-LDL-induced THP-1 macrophage-derived foam cells through modulating the promotion of cholesterol efflux and alleviation of inflammation. MATERIALS AND METHODS MTT was used to detect cell viability. Bodipy493/503 and oil red O staining were performed to measure cell lipid deposition. Enzymatic assay was used to measure intracellular cholesterol measurement. Cholesterol efflux/uptake were determined by cholesterol efflux assay and Dil-ox-LDL uptake assay. Inflammatory cytokines were measured by ELISA. Bioinformatics prediction and dual luciferase reporter assay were performed to validate miR-182-5p targeting HDAC9. Relative protein levels were evaluated by immunoblotting and relative gene levels were determined by quantitative real-time PCR. RESULTS Our results showed that GP-17 upregulated the expression of ABCA1, ABCG1 and miR-182-5p, but reduced HDAC9 expression levels in lipid-loaded macrophages, which promoted cholesterol efflux and inhibited lipid deposition. Additionally, GP-17 promoted the M2 phenotype of the macrophage and suppressed the inflammatory response in THP-1 macrophage-derived foam cells. Overexpression of HDAC9 or suppression of miR-182-5p eliminated the effects of ABCA1/G1 expression, lipid deposition and pro-inflammatory response. CONCLUSION These findings suggest that GP-17 exerts a beneficial effect on macrophage lipid deposition and inflammation responses through activating the miR-182-5p/HDAC9 signaling pathway.
Collapse
Affiliation(s)
- Wen-Yi Deng
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan, PR China
| | - Cheng-Long Zhou
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, Guangdong, PR China
| | - Meng-Ya Zeng
- Cardiovascular Disease Clinical Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan, PR China.
| |
Collapse
|
16
|
Chuecos MA, Lagor WR. Liver directed adeno-associated viral vectors to treat metabolic disease. J Inherit Metab Dis 2024; 47:22-40. [PMID: 37254440 PMCID: PMC10687323 DOI: 10.1002/jimd.12637] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/05/2023] [Accepted: 05/25/2023] [Indexed: 06/01/2023]
Abstract
The liver is the metabolic center of the body and an ideal target for gene therapy of inherited metabolic disorders (IMDs). Adeno-associated viral (AAV) vectors can deliver transgenes to the liver with high efficiency and specificity and a favorable safety profile. Recombinant AAV vectors contain only the transgene cassette, and their payload is converted to non-integrating circular double-stranded DNA episomes, which can provide stable expression from months to years. Insights from cellular studies and preclinical animal models have provided valuable information about AAV capsid serotypes with a high liver tropism. These vectors have been applied successfully in the clinic, particularly in trials for hemophilia, resulting in the first approved liver-directed gene therapy. Lessons from ongoing clinical trials have identified key factors affecting efficacy and safety that were not readily apparent in animal models. Circumventing pre-existing neutralizing antibodies to the AAV capsid, and mitigating adaptive immune responses to transduced cells are critical to achieving therapeutic benefit. Combining the high efficiency of AAV delivery with genome editing is a promising path to achieve more precise control of gene expression. The primary safety concern for liver gene therapy with AAV continues to be the small risk of tumorigenesis from rare vector integrations. Hepatotoxicity is a key consideration in the safety of neuromuscular gene therapies which are applied at substantially higher doses. The current knowledge base and toolkit for AAV is well developed, and poised to correct some of the most severe IMDs with liver-directed gene therapy.
Collapse
Affiliation(s)
- Marcel A. Chuecos
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX USA
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX USA
| | - William R. Lagor
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
17
|
Karpouzas GA, Papotti B, Ormseth SR, Palumbo M, Hernandez E, Adorni MP, Zimetti F, Budoff MJ, Ronda N. Inflammation and immunomodulatory therapies influence the relationship between ATP-binding cassette A1 membrane transporter-mediated cholesterol efflux capacity and coronary atherosclerosis in rheumatoid arthritis. J Transl Autoimmun 2023; 7:100209. [PMID: 37520890 PMCID: PMC10371792 DOI: 10.1016/j.jtauto.2023.100209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023] Open
Abstract
Objectives High-density lipoprotein (HDL) removes cholesterol from cells in atherosclerotic lesions, a function known as cholesterol efflux capacity (CEC). ATP-binding-cassette A1 (ABCA1) membrane transporter starts cholesterol transfer from macrophages to HDL particles. In rheumatoid arthritis (RA), methotrexate and biologic disease modifying drugs (bDMARDs) are atheroprotective whereas corticosteroids and C-reactive protein (CRP) are proatherogenic. We evaluated the influence of these factors on the relationship of ABCA1-CEC with atherosclerosis and cardiovascular events. Methods Atherosclerosis was evaluated with computed tomography angiography in 140 patients with RA and repeated in 99 after 6.9 ± 0.3 years. Events including acute coronary syndromes, stroke, cardiovascular death, claudication, revascularization, and heart failure were recorded. ABCA1-CEC was quantified in J774A.1 murine macrophages and reported as percentage of effluxed over intracellular cholesterol. Results Higher ABCA1-CEC associated with (i) more calcified plaques at baseline only in patients with CRP>7 mg/L (median) (p-interaction = 0.001) and methotrexate nonusers (p-interaction = 0.037), and more partially-calcified plaques only in bDMARD nonusers (p-interaction = 0.029); (ii) fewer new calcified plaques in patients with below-median but not higher time-averaged CRP (p-interaction = 0.028); (iii) fewer new total and calcified plaques in prednisone unexposed but not patients exposed to prednisone during follow-up (p-interaction = 0.034 and 0.004) and (iv) more new plaques in baseline bDMARD nonusers and fewer in bDMARD users (p-interaction ≤ 0.001). Also, ABCA1-CEC associated with greater cardiovascular risk only in baseline prednisone users (p-interaction = 0.027). Conclusion ABCA1-CEC associated with decreased atherosclerosis in patients with below-median baseline and time-averaged CRP and bDMARD use. Conversely, ABCA1-CEC associated with increased plaque in those with higher CRP, corticosteroid users, methotrexate nonusers, and bDMARD nonusers. While in well-treated and controlled disease ABCA1-CEC appears atheroprotective, in uncontrolled RA its action may be masked or fail to counteract the inflammation-driven proatherogenic state.
Collapse
Affiliation(s)
- George A. Karpouzas
- Division of Rheumatology, Harbor-UCLA and The Lundquist Institute, Torrance, CA, USA
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Sarah R. Ormseth
- Division of Rheumatology, Harbor-UCLA and The Lundquist Institute, Torrance, CA, USA
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | | | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Matthew J. Budoff
- Division of Cardiology, Harbor-UCLA and The Lundquist Institute, Torrance, CA, USA
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| |
Collapse
|
18
|
Karpouzas GA, Papotti B, Ormseth SR, Palumbo M, Hernandez E, Adorni MP, Zimetti F, Budoff MJ, Ronda N. Statins influence the relationship between ATP-binding cassette A1 membrane transporter-mediated cholesterol efflux capacity and coronary atherosclerosis in rheumatoid arthritis. J Transl Autoimmun 2023; 7:100206. [PMID: 37484708 PMCID: PMC10362327 DOI: 10.1016/j.jtauto.2023.100206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/23/2023] [Accepted: 07/04/2023] [Indexed: 07/25/2023] Open
Abstract
Objectives Cholesterol efflux capacity (CEC) is the main antiatherogenic function of high-density lipoprotein (HDL). ATP-binding-cassette A1 (ABCA1) membrane transporter initiates cholesterol export from arterial macrophages to pre-β HDL particles fostering their maturation; in turn, those accept cholesterol through ABCG1-mediated export. Impaired pre-β HDL maturation may disrupt the collaborative function of the two transporters and adversely affect atherosclerosis. Statins exert atheroprotective functions systemically and locally on plaque. We here evaluated associations between ABCA1-CEC, coronary atherosclerosis and cardiovascular risk and the influence of statins on those relationships in rheumatoid arthritis (RA). Methods Evaluation with computed tomography angiography was undertaken in 140 patients and repeated in 99 after 6.9 ± 0.3 years. Events comprising cardiovascular death, acute coronary syndromes, stroke, claudication, revascularization and heart failure were recorded. ABCA1-CEC and ABCG1-CEC were evaluated in J774A.1 macrophages and Chinese hamster ovary (CHO) cells respectively and expressed as percentage of effluxed over total intracellular cholesterol. Covariates in all cardiovascular event risk and plaque outcome models included atherosclerotic cardiovascular disease (ASCVD) risk score and high-density lipoprotein cholesterol. Results ABCA1-CEC negatively correlated with ABCG1-CEC (r = -0.167, p = 0.049). ABCA1-CEC associated with cardiovascular risk (adjusted hazard ratio 2.05 [95%CI 1.20-3.48] per standard deviation [SD] increment). There was an interaction of ABCA1-CEC with time-varying statin use (p = 0.038) such that current statin use inversely associated with risk only in patients with ABCA1-CEC below the upper tertile. ABCA1-CEC had no main effect on plaque or plaque progression; instead, ABCA1-CEC (per SD) associated with fewer baseline total plaques (adjusted rate ratio [aRR] 0.81, [95%CI 0.65-1.00]), noncalcified plaques (aRR 0.78 [95%CI 0.61-0.98]), and vulnerable low-attenuation plaques (aRR 0.41 [95%CI 0.23-0.74]) in statin users, and more low-attenuation plaques (aRR 1.91 [95%CI 1.18-3.08]) in nonusers (p-for-interaction = 0.018, 0.011, 0.025 and < 0.001 respectively). Moreover, ABCA1-CEC (per SD) associated with greater partially/fully-calcified plaque progression (adjusted odds ratio 3.07 [95%CI 1.20-7.86]) only in patients not exposed to statins during follow-up (p-for-interaction = 0.009). Conclusion In patients with RA, higher ABCA1-CEC may reflect a proatherogenic state, associated with enhanced cardiovascular risk. Statin use may unmask the protective impact of ABCA1-mediated cholesterol efflux on plaque formation, progression and cardiovascular risk.
Collapse
Affiliation(s)
- George A. Karpouzas
- Division of Rheumatology, Harbor-UCLA Medical Center and the Lundquist Institute for Biomedical Innovation, Torrance, CA, USA
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Sarah R. Ormseth
- Division of Rheumatology, Harbor-UCLA Medical Center and the Lundquist Institute for Biomedical Innovation, Torrance, CA, USA
| | | | - Elizabeth Hernandez
- Division of Rheumatology, Harbor-UCLA Medical Center and the Lundquist Institute for Biomedical Innovation, Torrance, CA, USA
| | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Matthew J. Budoff
- Division of Cardiology, Harbor-UCLA Medical Center and the Lundquist Institute for Biomedical Innovation, Torrance, CA, USA
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parma, Italy
| |
Collapse
|
19
|
Liu N, Tian J, Steer CJ, Han Q, Song G. MicroRNA-206-3p suppresses hepatic lipogenesis and cholesterol synthesis while driving cholesterol efflux. Hepatology 2023:01515467-990000000-00643. [PMID: 37943861 DOI: 10.1097/hep.0000000000000672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND AND AIMS Hepatosteatosis, hypertriglyceridemia, and hypercholesterolemia are interconnected metabolic disorders. This study is designed to characterize how microRNA-206-3p (miR-206) simultaneously prevents de novo lipogenesis (DNL), cholesterol synthesis, and VLDL production in hepatocytes while promoting cholesterol efflux in macrophages. APPROACH AND RESULTS MiR-206 levels were reduced in hepatocytes and macrophages of mice subjected to a high-fat, high-cholesterol diet. A negative feedback between LXRα (liver X receptor alpha) and miR-206 is formed to maintain high LXRα and low miR-206 in hepatocytes. Systemic administration of miR-206 alleviated hepatosteatosis, hypertriglyceridemia, and hypercholesterolemia in mice. A significant reduction in LDL cholesterol and VLDL cholesterol but unaltered HDL cholesterol was observed in miR-206-treated mice. Mirroring these findings, miR-206 reprogrammed the transcriptome of hepatocytes towards the inhibition of DNL, cholesterol synthesis, and assembly and secretion of VLDL. In macrophages, miR-206 activated the expression of genes regulating cholesterol efflux. Hepatocyte-specific expression of miR-206 reduced hepatic and circulating triglycerides and cholesterol, as well as VLDL production, while transplantation of macrophages bearing miR-206 facilitated cholesterol efflux. Mechanistically, miR-206 directly targeted Lxrα and Hmgcr in hepatocytes but facilitated expression of Lxrα in macrophages by targeting macrophage-specific tricho-rhino-phalangeal syndrome 1 (TRPS1), a transcription repressor of Lxrα . By targeting Hmgc r and Lxrα , miR-206 inhibited DNL, VLDL production, and cholesterol synthesis in hepatocytes, whereas it drove cholesterol efflux by activating the TRPS1-LXRα axis. CONCLUSIONS MiR-206, through differentially modulating LXRα signaling in hepatocytes and macrophages, inhibits DNL, promotes cholesterol efflux, and concurrently hinders cholesterol synthesis and VLDL production. MiR-206 simulates the functions of lipid-lowering medications, statins, and LXRα agonists.
Collapse
Affiliation(s)
- Ningning Liu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Jing Tian
- Department of Cardiology, the First Hospital of Shanxi Medical University, Taiyuan City, China
| | - Clifford J Steer
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Qinghua Han
- Department of Cardiology, the First Hospital of Shanxi Medical University, Taiyuan City, China
| | - Guisheng Song
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
20
|
He Y, Pavanello C, Hutchins PM, Tang C, Pourmousa M, Vaisar T, Song HD, Pastor RW, Remaley AT, Goldberg IJ, Costacou T, Davidson WS, Bornfeldt KE, Calabresi L, Segrest JP, Heinecke JW. Flipped C-Terminal Ends of APOA1 Promote ABCA1-dependent Cholesterol Efflux by Small HDLs. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.03.23297986. [PMID: 37961344 PMCID: PMC10635269 DOI: 10.1101/2023.11.03.23297986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Cholesterol efflux capacity (CEC) predicts cardiovascular disease (CVD) independently of HDL cholesterol (HDL-C) levels. Isolated small HDL particles are potent promoters of macrophage CEC by the ABCA1 pathway, but the underlying mechanisms are unclear. Methods We used model system studies of reconstituted HDL and plasma from control and lecithin-cholesterol acyltransferase (LCAT)-deficient subjects to investigate the relationships among the sizes of HDL particles, the structure of APOA1 in the different particles, and the CECs of plasma and isolated HDLs. Results We quantified macrophage and ABCA1 CEC of four distinct sizes of reconstituted HDL (r-HDL). CEC increased as particle size decreased. MS/MS analysis of chemically crosslinked peptides and molecular dynamics simulations of APOA1 (HDL's major protein) indicated that the mobility of that protein's C-terminus was markedly higher and flipped off the surface in the smallest particles. To explore the physiological relevance of the model system studies, we isolated HDL from LCAT-deficient subjects, whose small HDLs-like r-HDLs-are discoidal and composed of APOA1, cholesterol, and phospholipid. Despite their very low plasma levels of HDL particles, these subjects had normal CEC. In both the LCAT-deficient subjects and control subjects, the CEC of isolated extra-small HDL (a mixture of extra-small and small HDL by calibrated ion mobility analysis) was 3-5-fold greater than that of the larger sizes of isolated HDL. Incubating LCAT-deficient plasma and control plasma with human LCAT converted extra-small and small HDL particles into larger particles, and it markedly inhibited CEC. Conclusions We present a mechanism for the enhanced CEC of small HDLs. In smaller particles, the C-termini of the two antiparallel molecules of APOA1 are flipped off the lipid surface of HDL. This extended conformation allows them to engage with ABCA1. In contrast, the C-termini of larger HDLs are unable to interact productively with ABCA1 because they form a helical bundle that strongly adheres to the lipid on the particle. Enhanced CEC, as seen with the smaller particles, predicts decreased CVD risk. Thus, extra-small and small HDLs may be key mediators and indicators of HDL's cardioprotective effects.
Collapse
Affiliation(s)
- Yi He
- Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Chiara Pavanello
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Patrick M Hutchins
- Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Chongren Tang
- Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Mohsen Pourmousa
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Tomas Vaisar
- Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Hyun D Song
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240, USA
| | - Richard W Pastor
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Alan T Remaley
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD 20892
| | - Ira J Goldberg
- Department of Medicine, New York University, New York, NY, 10016, USA
| | - Tina Costacou
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45237, USA
| | - Karin E Bornfeldt
- Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Laura Calabresi
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Jere P Segrest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240, USA
| | - Jay W Heinecke
- Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| |
Collapse
|
21
|
Marghani BH, Ateya AI, Othman BH, Rizk MA, El-Adl M. UGT1A1 morpholino antisense oligonucleotides produce mild unconjugated hyperbilirubinemia in cyclosporine A-induced cardiovascular disorders in BLC57 mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 104:104321. [PMID: 37984676 DOI: 10.1016/j.etap.2023.104321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 11/11/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
This study aimed to investigate the induction of mild unconjugated hyperbilirubinemia in hepatic UGT1A1 inhibition by Morpholinos antisense in CsA-treated BLC57 mice in comparison with the efficacy of chitosan (CH) as an anti-hypolipidemic natural product. Antisense morpholino oligonucleotides were injected intravenously into CsA-treated mice for 14 days thrice a week. Serum biochemical parameters, antioxidant status, and gene expression analysis of eNOS, PPAR-α, NF-kB, cFn, AT1-R, and ETA-R were determined in cardiac tissues with confirmation by histopathology. Inhibition of UGT1A1 significantly elevated serum unconjugated bilirubin within a physiological range. Furthermore, induced mild hyperbilirubinemia reduces hyperlipidemia, improves antioxidant status, and significantly increases the expression of the cardiac PPAR-α gene while decreasing, ETA-R, iNOS, NF-kB, cFn and AT1-R gene expression in CsA-treated mice. Importantly, mild unconjugated hyperbilirubinemia within physiological ranges may be used as a novel therapeutic strategy to lower hyperlipidemia, atherosclerosis, hypertension, and the CVD outcomes in CsA- treated transplant recipients.
Collapse
Affiliation(s)
- Basma H Marghani
- Department of Biochemistry, Physiology, and Pharmacology, Faculty of Veterinary Medicine, King Salman International University, South of Sinai 46612, Egypt; Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed I Ateya
- Department of Husbandry & Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Basma H Othman
- Medical Experimental Research Center, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed Abdo Rizk
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed El-Adl
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
22
|
Feng RQ, Xu MY, Feng RY, Zhang L, Yin XF, Wang C, Liu JB. Serum Isthmin-1 is negatively correlated with HDL-C in type 2 diabetes mellitus. J Diabetes Complications 2023; 37:108567. [PMID: 37647712 DOI: 10.1016/j.jdiacomp.2023.108567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Isthmin-1 (Ism-1) is a newly identified insulin-like adipokine that increases glucose uptake by adipocytes and inhibits hepatic lipid synthesis. Recent studies have shown that Ism-1 can improve the metabolic disorders associated with type 2 diabetes mellitus (T2DM) and improve lipid metabolism. The classic function of high-density lipoprotein cholesterol (HDL-C) is to transport cholesterol from extra-hepatic tissues to the liver for metabolism. In contrast, disorders of lipid metabolism and inflammation are the leading causes of atherosclerosis (As). Atherosclerosis often manifests as loss of elasticity, lipid accumulation, fibrous tissue proliferation and calcium deposits in the affected arteries, eventually forming plaques. AIM To illustrate the correlation between HDL-C and Ism-1 in T2DM, and the relationship between lipoprotein cholesterol and carotid plaque. METHODS A total of 128 patients with T2DM were enrolled in the study and basic information was collected. HDL-C levels were measured chemically. Serum Ism-1 levels were measured using an enzyme-linked immunosorbent assay (ELISA). Linear regression analysis was used to assess the correlation between serum Ism-1 levels and HDL-C in patients with T2DM. Basic information was again collected from 226 patients with T2DM. Independent sample t-tests were performed to explore the relationship between carotid plaque formation and lipids. RESULTS HDL-C was divided into four groups according to quartiles and there was a between-group difference in Ism-1 (p = 0.040). Multivariable linear regression showed a negative association between Ism-1 and HDL-C in T2DM (β = -0.235, p < 0.001), even after adjusting for related factors (β = -0.165, p = 0.009). Low-density lipoprotein cholesterol (LDL-C) and HDL-C showed significant differences between the carotid plaque group and the non-carotid plaque group (pLDL-C = 0.007, pHDL-C = 0.003). CONCLUSION Serum Ism-1 and HDL-C are negatively correlated in T2DM. LDL-C is significantly higher in carotid plaque group than non-carotid plaque group, while HDL-C is significantly lower than in the non-carotid plaque group.
Collapse
Affiliation(s)
- Ruo-Qi Feng
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Ming-Yue Xu
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Rui-Ying Feng
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Lei Zhang
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Xiao-Fei Yin
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Chuan Wang
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Jin-Bo Liu
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China; Institute of Endocrine and Metabolic Diseases, Shandong University, Jinan 250012, Shandong Province, China.
| |
Collapse
|
23
|
Bi L, Wacker BK, Komandur K, Sanford N, Dichek DA. Apolipoprotein A-I vascular gene therapy reduces vein-graft atherosclerosis. Mol Ther Methods Clin Dev 2023; 30:558-572. [PMID: 37693942 PMCID: PMC10482902 DOI: 10.1016/j.omtm.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023]
Abstract
Coronary artery venous bypass grafts typically fail because of atherosclerosis driven by lipid and macrophage accumulation. Therapy for vein-graft atherosclerosis is limited to statin drugs, which are only modestly effective. We hypothesized that transduction of vein-graft endothelium of fat-fed rabbits with a helper-dependent adenovirus expressing apolipoprotein AI (HDAdApoAI) would reduce lipid and macrophage accumulation. Fat-fed rabbits received bilateral external jugular vein-to-carotid artery interposition grafts. Four weeks later, one graft per rabbit (n = 23 rabbits) was infused with HDAdApoAI and the contralateral graft with HDAdNull. Grafts were harvested 12 weeks later. Paired analyses of grafts were performed, with vein graft cholesterol, intimal lipid, and macrophage content as the primary endpoints. HDAd genomes were detected in all grafts. APOAI mRNA was median 63-fold higher in HDAdApoAI grafts versus HDAdNull grafts (p < 0.001). HDAdApoAI grafts had a mean 15% lower total cholesterol (by mass spectrometry; p = 0.003); mean 19% lower intimal lipid (by oil red O staining; p = 0.02); and mean 13% lower expression of the macrophage marker CD68 (by reverse transcriptase-mediated quantitative PCR; p = 0.008). In vivo transduction of vein-graft endothelium achieves persistent APOAI expression and reduces vein-graft cholesterol, intimal lipid, and CD68 expression. Vascular gene therapy with APOAI has promise for preventing vein-graft failure caused by atherosclerosis.
Collapse
Affiliation(s)
- Lianxiang Bi
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Bradley K. Wacker
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Kaushik Komandur
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Nicole Sanford
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - David A. Dichek
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| |
Collapse
|
24
|
Darabi M, Lhomme M, Ponnaiah M, Pučić-Baković M, Guillas I, Frisdal E, Bittar R, Croyal M, Matheron-Duriez L, Poupel L, Bonnefont-Rousselot D, Frere C, Varret M, Krempf M, Cariou B, Lauc G, Guerin M, Carrie A, Bruckert E, Giral P, Le Goff W, Kontush A. Integrated omics approach for the identification of HDL structure-function relationships in PCSK9-related familial hypercholesterolemia. J Clin Lipidol 2023; 17:643-658. [PMID: 37550151 DOI: 10.1016/j.jacl.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND The role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in dyslipidemia may go beyond its immediate effects on low-density lipoprotein receptor (LDL-R) activity. OBJECTIVE This study aimed to assess PCSK9-derived alterations of high-density lipoprotein (HDL) physiology, which bear a potential to contribute to cardiovascular risk profile. METHODS HDL was isolated from 33 patients with familial autosomal dominant hypercholesterolemia (FH), including those carrying PCSK9 gain-of-function (GOF) genetic variants (FH-PCSK9, n = 11), together with two groups of dyslipidemic patients employed as controls and carrying genetic variants in the LDL-R not treated (ntFH-LDLR, n = 11) and treated (tFH-LDLR, n = 11) with statins, and 11 normolipidemic controls. Biological evaluations paralleled by proteomic, lipidomic and glycomic analyses were applied to characterize functional and compositional properties of HDL. RESULTS Multiple deficiencies in the HDL function were identified in the FH-PCSK9 group relative to dyslipidemic FH-LDLR patients and normolipidemic controls, which involved reduced antioxidative, antiapoptotic, anti-thrombotic and anti-inflammatory activities. By contrast, cellular cholesterol efflux capacity of HDL was unchanged. In addition, multiple alterations of the proteomic, lipidomic and glycomic composition of HDL were found in the FH-PCSK9 group. Remarkably, HDLs from FH-PCSK9 patients were systematically enriched in several lysophospholipids as well as in A2G2S2 (GP13) glycan and apolipoprotein A-IV. Based on network analysis of functional and compositional data, a novel mosaic structure-function model of HDL biology involving FH was developed. CONCLUSION Several metrics of anti-atherogenic HDL functionality are altered in FH-PCSK9 patients paralleled by distinct compositional alterations. These data provide a first-ever overview of the impact of GOF PCSK9 genetic variants on structure-function relationships in HDL.
Collapse
Affiliation(s)
- Maryam Darabi
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France; LPS-BioSciences (Current affiliation of Dr Darabi), Université de Paris-Saclay, Orsay, France
| | - Marie Lhomme
- ICAN Analytics (Dr Lhomme), Lipidomics Core, Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), Paris, France
| | - Maharajah Ponnaiah
- ICAN I/O (Dr Ponnaiah), Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), Paris, France
| | - Maja Pučić-Baković
- Genos Glycoscience Research Laboratory (Drs Pučić-Baković and Lauc), Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| | - Isabelle Guillas
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Eric Frisdal
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Randa Bittar
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France; Department of Metabolic Biochemistry (Drs Bittar and Bonnefont-Rousselot), Pitié-Salpêtrière-Charles Foix Hospital, AP-HP, Paris, France
| | - Mikaël Croyal
- Université de Nantes (Drs Cariou et Croyal), CHU Nantes, CNRS, INSERM, l'Institut du Thorax, F-44000 Nantes, France; Université de Nantes (Dr Croyal), CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, F-44000 Nantes, France; CRNH-Ouest Mass Spectrometry Core Facility (Drs Croyal and Krempf), F-44000 Nantes, France
| | - Lucrèce Matheron-Duriez
- Platform MS3U (Dr Matheron), Institut de Biologie Paris Seine FR 3631, Sorbonne Université, Paris, France
| | - Lucie Poupel
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Dominique Bonnefont-Rousselot
- Department of Metabolic Biochemistry (Drs Bittar and Bonnefont-Rousselot), Pitié-Salpêtrière-Charles Foix Hospital, AP-HP, Paris, France; Université de Paris (Dr Bonnefont-Rousselot), CNRS, INSERM, UTCBS, F-75006 Paris, France
| | - Corinne Frere
- Department of Haematology (Dr Frere), Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Mathilde Varret
- Paris University and Sorbonne Paris Nord University (Dr Varret), National Institute for Health and Medical Research (INSERM, LVTS), F-75018 Paris, France
| | - Michel Krempf
- CRNH-Ouest Mass Spectrometry Core Facility (Drs Croyal and Krempf), F-44000 Nantes, France; Clinique Bretéché (Dr Krempf), Groupe Elsan, Nantes, France
| | - Bertrand Cariou
- Université de Nantes (Drs Cariou et Croyal), CHU Nantes, CNRS, INSERM, l'Institut du Thorax, F-44000 Nantes, France
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory (Drs Pučić-Baković and Lauc), Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| | - Maryse Guerin
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Alain Carrie
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Eric Bruckert
- Endocrinologie Métabolisme et Prévention Cardiovasculaire (Drs Bruckert and Giral), Institut E3M et IHU Cardiométabolique (ICAN), Hôpital Pitié Salpêtrière, Paris, France
| | - Philippe Giral
- Endocrinologie Métabolisme et Prévention Cardiovasculaire (Drs Bruckert and Giral), Institut E3M et IHU Cardiométabolique (ICAN), Hôpital Pitié Salpêtrière, Paris, France
| | - Wilfried Le Goff
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France
| | - Anatol Kontush
- Sorbonne Université, INSERM (Drs Darabi, Guillas, Frisdal, Poupel, Carrie,Bittar, Guerin, Le Goff, and Kontush), Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, F-75013 Paris, France.
| |
Collapse
|
25
|
Angeli V, Lim HY. Biomechanical control of lymphatic vessel physiology and functions. Cell Mol Immunol 2023; 20:1051-1062. [PMID: 37264249 PMCID: PMC10469203 DOI: 10.1038/s41423-023-01042-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/26/2023] [Accepted: 04/29/2023] [Indexed: 06/03/2023] Open
Abstract
The ever-growing research on lymphatic biology has clearly identified lymphatic vessels as key players that maintain human health through their functional roles in tissue fluid homeostasis, immunosurveillance, lipid metabolism and inflammation. It is therefore not surprising that the list of human diseases associated with lymphatic malfunctions has grown larger, including issues beyond lymphedema, a pathology traditionally associated with lymphatic drainage insufficiency. Thus, the discovery of factors and pathways that can promote optimal lymphatic functions may offer new therapeutic options. Accumulating evidence indicates that aside from biochemical factors, biomechanical signals also regulate lymphatic vessel expansion and functions postnatally. Here, we review how mechanical forces induced by fluid shear stress affect the behavior and functions of lymphatic vessels and the mechanisms lymphatic vessels employ to sense and transduce these mechanical cues into biological signals.
Collapse
Affiliation(s)
- Veronique Angeli
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore.
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
26
|
Iyer DR, Venkatraman J, Tanguy E, Vitale N, Mahapatra NR. Chromogranin A and its derived peptides: potential regulators of cholesterol homeostasis. Cell Mol Life Sci 2023; 80:271. [PMID: 37642733 PMCID: PMC11072126 DOI: 10.1007/s00018-023-04908-3] [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: 03/03/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023]
Abstract
Chromogranin A (CHGA), a member of the granin family of proteins, has been an attractive therapeutic target and candidate biomarker for several cardiovascular, neurological, and inflammatory disorders. The prominence of CHGA stems from the pleiotropic roles of several bioactive peptides (e.g., catestatin, pancreastatin, vasostatins) generated by its proteolytic cleavage and by their wide anatomical distribution. These peptides are emerging as novel modulators of cardiometabolic diseases that are often linked to high blood cholesterol levels. However, their impact on cholesterol homeostasis is poorly understood. The dynamic nature of cholesterol and its multitudinous roles in almost every aspect of normal body function makes it an integral component of metabolic physiology. A tightly regulated coordination of cholesterol homeostasis is imperative for proper functioning of cellular and metabolic processes. The deregulation of cholesterol levels can result in several pathophysiological states. Although studies till date suggest regulatory roles for CHGA and its derived peptides on cholesterol levels, the mechanisms by which this is achieved still remain unclear. This review aims to aggregate and consolidate the available evidence linking CHGA with cholesterol homeostasis in health and disease. In addition, we also look at common molecular regulatory factors (viz., transcription factors and microRNAs) which could govern the expression of CHGA and genes involved in cholesterol homeostasis under basal and pathological conditions. In order to gain further insights into the pathways mediating cholesterol regulation by CHGA/its derived peptides, a few prospective signaling pathways are explored, which could act as primers for future studies.
Collapse
Affiliation(s)
- Dhanya R Iyer
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Janani Venkatraman
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Emeline Tanguy
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France.
| | - Nitish R Mahapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.
| |
Collapse
|
27
|
Noflatscher M, Hunjadi M, Schreinlechner M, Sommer P, Lener D, Theurl M, Kirchmair R, Bauer A, Ritsch A, Marschang P. Inverse Correlation of Cholesterol Efflux Capacity with Peripheral Plaque Volume Measured by 3D Ultrasound. Biomedicines 2023; 11:1918. [PMID: 37509557 PMCID: PMC10376979 DOI: 10.3390/biomedicines11071918] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
INTRODUCTION Cardiovascular disease (CVD) is a systemic multifocal illness called atherosclerosis that causes artery constriction and blockage. By causing cholesterol to build up in the artery wall, hypercholesterolemia is a major factor in the pathophysiology of atherosclerotic plaque development. Reverse cholesterol transport is the process of transporting cholesterol from the periphery back to the liver through cholesterol efflux mediated by high-density lipoprotein (HDL). It was suggested that the cholesterol efflux capacity (CEC), which is inversely linked with cardiovascular risk, can serve as a stand-in measure for reverse cholesterol transport. In this work, we sought to investigate a potential link between the peripheral plaque volume (PV) and CEC. METHODS Since lipid-lowering therapy interferes with CEC, we performed a cross-sectional study of 176 patients (48.9% females) with one cardiovascular risk factor or known CVD that did not currently take lipid-lowering medication. CEC was determined using cAMP-treated 3H-cholesterol-labeled J774 cells. Cholesterol ester transfer protein (CETP)-mediated cholesterol ester transfer was measured by quantifying the transfer of cholesterol ester from radiolabeled exogenous HDL cholesterol to Apolipoprotein B-containing lipoproteins. PV in the carotid and the femoral artery, defined as the total PV, was measured using a 3D ultrasound system equipped with semi-automatic software. RESULTS In our patients, we discovered an inverse relationship between high total PV and CEC (p = 0.027). However, there was no connection between total PV and low-density lipoprotein cholesterol, lipoprotein (a), or CETP-mediated cholesterol ester transfer. CONCLUSION In patients not receiving lipid-lowering treatment, CEC inversely correlates with peripheral atherosclerosis, supporting its role in the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Maria Noflatscher
- Department of Internal Medicine III (Cardiology, Angiology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Monika Hunjadi
- Department of Internal Medicine I, Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Michael Schreinlechner
- Department of Internal Medicine III (Cardiology, Angiology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Philip Sommer
- Department of Internal Medicine III (Cardiology, Angiology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Daniela Lener
- Department of Internal Medicine III (Cardiology, Angiology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Markus Theurl
- Department of Internal Medicine III (Cardiology, Angiology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Rudolf Kirchmair
- Department of Internal Medicine III (Cardiology, Angiology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Axel Bauer
- Department of Internal Medicine III (Cardiology, Angiology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Andreas Ritsch
- Department of Internal Medicine I, Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | - Peter Marschang
- Department of Internal Medicine III (Cardiology, Angiology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
- Department of Internal Medicine, Central Hospital of Bolzano (SABES-ASDAA), Via Lorenz Boehler 5, I-39100 Bolzano, Italy
| |
Collapse
|
28
|
Shi X, Feng D, Li D, Han P, Yang L, Wei W. A pan-cancer analysis of the oncogenic and immunological roles of apolipoprotein F (APOF) in human cancer. Eur J Med Res 2023; 28:190. [PMID: 37312170 DOI: 10.1186/s40001-023-01156-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/03/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Apolipoprotein F (APOF) has been less studied in cancers. Thus, we aimed to perform a pan-cancer analysis of the oncogenic and immunological effects of APOF on human cancer. METHODS A standardized TCGA pan-cancer dataset was downloaded. Differential expression, clinical prognosis, genetic mutations, immune infiltration, epigenetic modifications, tumor stemness and heterogeneity were analyzed. We conducted all analyses through software R (version 3.6.3) and its suitable packages. RESULTS Overall, we found that the common cancers differentially expressed between tumor and normal samples and prognostic-associated were BRCA, PRAD, KIRP, and LIHC in terms of overall survival (OS), disease-free survival (DFS) and progression-free survival (PFS). The pan-cancer Spearman analysis showed that the mRNA expression of APOF was negatively correlated with four tumor stemness indexes (DMPss, DNAss, ENHss, and EREG-METHss) with statistical significance for PRAD and was positively correlated for LIHC. In terms of BRCA and PRAD patients, we found negative correlation of APOF with TMB, MSI, neo, HRD and LOH. The mutation frequencies of BRCA and LIHC were 0.3%. APOF expression was negatively correlated with immune infiltration and positively correlated with tumor purity for PRAD patients. The mRNA expression of APOF was negatively associated with most TILs for LIHC, B cells, CD4+ T cells, neutrophils, macrophages and dendritic cells, but was positively associated with CD8+ T cells. CONCLUSIONS Our pan-cancer study offered a relatively comprehensive understanding of the roles of APOF on BRCA, PRAD, KIRP, and LIHC.
Collapse
Affiliation(s)
- Xu Shi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, China
| | - Ping Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, China
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Guoxue Xiang #37, Chengdu, 610041, China.
| |
Collapse
|
29
|
de Oliveira Laterza Ribeiro M, Correia VM, Herling de Oliveira LL, Soares PR, Scudeler TL. Evolving Diagnostic and Management Advances in Coronary Heart Disease. Life (Basel) 2023; 13:951. [PMID: 37109480 PMCID: PMC10143565 DOI: 10.3390/life13040951] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/02/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Despite considerable improvement in diagnostic modalities and therapeutic options over the last few decades, the global burden of ischemic heart disease is steadily rising, remaining a major cause of death worldwide. Thus, new strategies are needed to lessen cardiovascular events. Researchers in different areas such as biotechnology and tissue engineering have developed novel therapeutic strategies such as stem cells, nanotechnology, and robotic surgery, among others (3D printing and drugs). In addition, advances in bioengineering have led to the emergence of new diagnostic and prognostic techniques, such as quantitative flow ratio (QFR), and biomarkers for atherosclerosis. In this review, we explore novel diagnostic invasive and noninvasive modalities that allow a more detailed characterization of coronary disease. We delve into new technological revascularization procedures and pharmacological agents that target several residual cardiovascular risks, including inflammatory, thrombotic, and metabolic pathways.
Collapse
Affiliation(s)
| | | | | | | | - Thiago Luis Scudeler
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| |
Collapse
|
30
|
Karpouzas GA, Papotti B, Ormseth SR, Palumbo M, Hernandez E, Adorni MP, Zimetti F, Budoff MJ, Ronda N. ATP-binding cassette G1 membrane transporter-mediated cholesterol efflux capacity influences coronary atherosclerosis and cardiovascular risk in Rheumatoid Arthritis. J Autoimmun 2023; 136:103029. [PMID: 36996698 DOI: 10.1016/j.jaut.2023.103029] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
OBJECTIVES Cholesterol efflux capacity (CEC) measures the ability of high-density lipoprotein (HDL) to remove cholesterol from macrophages and reduce the lipid content of atherosclerotic plaques. CEC inversely associated with cardiovascular risk beyond HDL-cholesterol levels. CEC through the ATP-binding-cassette G1 (ABCG1) membrane transporter is impaired in rheumatoid arthritis (RA). We evaluated associations of ABCG1-CEC with coronary atherosclerosis, plaque progression and cardiovascular risk in RA. METHODS Coronary atherosclerosis (noncalcified, partially, fully-calcified, low-attenuation plaque) was assessed with computed tomography angiography in 140 patients and reevaluated in 99 after 6.9 ± 0.3 years. Cardiovascular events including acute coronary syndromes, stroke, cardiovascular death, claudication, revascularization and hospitalized heart failure were recorded. ABCG1-CEC was measured in Chinese hamster ovary cells as percentage of effluxed over total intracellular cholesterol. RESULTS ABCG1-CEC inversely associated with extensive atherosclerosis (≥5 plaques) (adjusted odds ratio 0.50 [95% CI 0.28-0.88]), numbers of partially-calcified (rate ratio [RR] 0.71 [0.53-0.94]) and low-attenuation plaques (RR 0.63 [0.43-0.91] per standard deviation increment). Higher ABCG1-CEC predicted fewer new partially-calcified plaques in patients with lower baseline and time-averaged CRP and fewer new noncalcified and calcified plaques in those receiving higher mean prednisone dose. ABCG1-CEC inversely associated with events in patients with but not without noncalcified plaques, with <median but not higher CRP and in prednisone users but not nonusers (p-for-interaction = 0.021, 0.033 and 0.008 respectively). CONCLUSION ABCG1-CEC inversely associated with plaque burden and vulnerability, and plaque progression conditionally on cumulative inflammation and corticosteroid dose. ABCG1-CEC inversely associated with events specifically in patients with noncalcified plaques, lower inflammation and in prednisone users.
Collapse
Affiliation(s)
- George A Karpouzas
- Division of Rheumatology, Harbor-UCLA Medical Center and the Lundquist Institute for Biomedical Innovation, Torrance, CA, USA.
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Sarah R Ormseth
- Division of Rheumatology, Harbor-UCLA Medical Center and the Lundquist Institute for Biomedical Innovation, Torrance, CA, USA
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Elizabeth Hernandez
- Division of Rheumatology, Harbor-UCLA Medical Center and the Lundquist Institute for Biomedical Innovation, Torrance, CA, USA
| | - Maria Pia Adorni
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Matthew J Budoff
- Division of Cardiology, Harbor-UCLA Medical Center and the Lundquist Institute for Biomedical Innovation, Torrance, CA, USA
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| |
Collapse
|
31
|
Kim HJ, Jeong S, Oh YH, Park SJ, Cho Y, Park SM. Changes in high-density lipoprotein cholesterol with risk of Cardiovascular Disease among initially high-density lipoprotein-high participants. Cardiovasc Diabetol 2023; 22:71. [PMID: 36978181 PMCID: PMC10053475 DOI: 10.1186/s12933-023-01805-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND High-density lipoprotein cholesterol's (HDL-C) long-held status as a cardiovascular disease (CVD) preventative has been called into question. Most of the evidence, however, focused on either the risk of death from CVD, or on single time point level of HDL-C. This study aimed to determine the association between changes in HDL-C levels and incident CVD in individuals with high baseline HDL-C levels (≥ 60 mg/dL). METHODS 77,134 people from the Korea National Health Insurance Service-Health Screening Cohort were followed for 517,515 person-years. Cox proportional hazards regression was used to evaluate the association between change in HDL-C levels and the risk of incident CVD. All participants were followed up until 31 December 2019, CVD, or death. RESULTS Participants with the greatest increase in their HDL-C levels had higher risks of CVD (adjusted hazard ratio [aHR], 1.15; 95% confidence interval [CI], 1.05-1.25) and CHD (aHR 1.27, CI 1.11-1.46) after adjusting for age, sex, household income, body mass index, hypertension, diabetes mellitus, dyslipidemia, smoking, alcohol consumption, moderate-to-vigorous physical activity, Charlson comorbidity index, and total cholesterol than those with the lowest increase in HDL-C levels. Such association remained significant even among participants with decreased low-density lipoprotein cholesterol (LDL-C) levels for CHD (aHR 1.26, CI 1.03-1.53). CONCLUSIONS In people with already high HDL-C levels, additional increases in HDL-C levels may be associated with an increased risk of CVD. This finding held true irrespective of the change in their LDL-C levels. Increasing HDL-C levels may lead to unintentionally elevated risk of CVD.
Collapse
Affiliation(s)
- Hye Jun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics, CHA University School of Medicine, Seongnam, South Korea
| | - Yun Hwan Oh
- Department of Family Medicine, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong, Korea
| | - Sun Jae Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoosun Cho
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Min Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Family Medicine, Seoul National University Hospital, Seoul, South Korea.
| |
Collapse
|
32
|
Gasbarrino K, Hafiane A, Gianopoulos I, Zheng H, Mantzoros CS, Daskalopoulou SS. Relationship between circulating adipokines and cholesterol efflux in subjects with severe carotid atherosclerosis. Metabolism 2023; 140:155381. [PMID: 36566801 DOI: 10.1016/j.metabol.2022.155381] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/02/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
AIMS Cholesterol efflux capacity (CEC) as a measure of high-density lipoprotein functionality is independently and inversely associated with increased risk of cardiovascular events and mortality, and advanced plaque morphology. Adipokines, adipose tissue-derived factors, can influence systemic lipoprotein metabolism, and participate in the regulation of vascular function and inflammation. We aimed to investigate the association between CEC and circulating adipokine levels (anti-inflammatory adiponectin, and pro-inflammatory chemerin and resistin) in subjects with severe carotid atherosclerotic disease and evaluate its impact on post-surgical outcomes. METHODS AND RESULTS This is a cross-sectional study with a 5-year follow-up component. Consecutive patients with severe carotid atherosclerosis scheduled for a carotid endarterectomy were recruited from hospital-based centres in Montreal, Canada (n = 285). Fasting blood samples were collected pre-operatively and used to measure plasma total and high-molecular weight (HMW) adiponectin, chemerin, and resistin, and to perform cholesterol efflux assays in J774 macrophage-like cells. Five-year post-surgery outcomes were obtained through medical chart review. Subjects had a mean age of 70.1 ± 9.4, were 67.0 % male, had various comorbidities (hypercholesterolemia [85.3 %], hypertension [83.5 %], type 2 diabetes [34.5 %], coronary artery disease [38.6 %]), and previously experienced cerebrovascular symptomatology (77.9 %). CEC was independently and positively associated with total and HMW adiponectin levels (ß [95 % confidence interval]; 0.216 [0.134-0.298] and 0.107 [0.037-0.176], respectively) but not with chemerin or resistin. Total adiponectin had the greatest association accounting for 8.3 % of the variance in CEC. Interaction regression models demonstrated a significant interaction between adiponectin and chemerin in increasing CEC. Notably, with each unit increase in CEC there was a 93.9 % decrease in the odds of having an ischemic cerebrovascular event 5 years post-surgery (0.061 [0.007-0.561]). CONCLUSIONS Our findings demonstrated circulating adiponectin to have a strong association with increased CEC in subjects with severe carotid atherosclerosis and high CEC to be associated with more favourable post-surgical outcomes. These findings reflect the importance of adipose tissue health in influencing CEC levels and atherosclerotic cardiovascular disease risk.
Collapse
Affiliation(s)
- Karina Gasbarrino
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada
| | - Anouar Hafiane
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada
| | - Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada
| | - Huaien Zheng
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA, United States
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University Montreal, Canada; Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, McGill University Montreal, Canada.
| |
Collapse
|
33
|
Electronegative LDL Is Associated with Plaque Vulnerability in Patients with Ischemic Stroke and Carotid Atherosclerosis. Antioxidants (Basel) 2023; 12:antiox12020438. [PMID: 36829998 PMCID: PMC9952764 DOI: 10.3390/antiox12020438] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Owing to the high risk of recurrence, identifying indicators of carotid plaque vulnerability in atherothrombotic ischemic stroke is essential. In this study, we aimed to identify modified LDLs and antioxidant enzymes associated with plaque vulnerability in plasma from patients with a recent ischemic stroke and carotid atherosclerosis. Patients underwent an ultrasound, a CT-angiography, and an 18F-FDG PET. A blood sample was obtained from patients (n = 64, 57.8% with stenosis ≥50%) and healthy controls (n = 24). Compared to the controls, patients showed lower levels of total cholesterol, LDL cholesterol, HDL cholesterol, apolipoprotein B (apoB), apoA-I, apoA-II, and apoE, and higher levels of apoJ. Patients showed lower platelet-activating factor acetylhydrolase (PAF-AH) and paraoxonase-1 (PON-1) enzymatic activities in HDL, and higher plasma levels of oxidized LDL (oxLDL) and electronegative LDL (LDL(-)). The only difference between patients with stenosis ≥50% and <50% was the proportion of LDL(-). In a multivariable logistic regression analysis, the levels of LDL(-), but not of oxLDL, were independently associated with the degree of carotid stenosis (OR: 5.40, CI: 1.15-25.44, p < 0.033), the presence of hypoechoic plaque (OR: 7.52, CI: 1.26-44.83, p < 0.027), and of diffuse neovessels (OR: 10.77, CI: 1.21-95.93, p < 0.033), indicating that an increased proportion of LDL(-) is associated with vulnerable atherosclerotic plaque.
Collapse
|
34
|
Development and validation of novel automatable assay for cholesterol efflux capacity. Biosci Rep 2023; 43:232455. [PMID: 36645426 PMCID: PMC9905788 DOI: 10.1042/bsr20221519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/24/2022] [Accepted: 01/16/2023] [Indexed: 01/17/2023] Open
Abstract
During the past decade, evaluation of high-density lipoprotein (HDL) functionality has been well studied for predicting cardiovascular disease (CVD) risk. Cholesterol efflux capacity (CEC) is the strongest candidate as the biomarker out of various HDL antiatherosclerotic functions. However, CEC has not yet been introduced clinically because of several technical issues, including the use of radioactive materials and differentiated cells in the assay. Previously, our laboratory developed a radioisotope- and cell-free CEC assay called the immobilized liposome-bound gel beads (ILGs) method to replace the conventional method. However, the separation process of the supernatant was not suitable for installation in an automatic analyzer. The present study aims to develop a new method that is easier to operate. We assumed that the use of magnetic beads instead of gel beads would enable the skip of the centrifugal process. First, similar to the ILG method, porous magnetic beads were treated with liposomes containing fluorescently labeled cholesterol. Fluorescence was observed inside the magnetic beads, and almost the same amount of liposomes as in the ILG method was immobilized successfully. These immobilized liposome-bound magnetic beads (ILMs) were available for CEC assay when HDL and apolipoprotein B-100-depleted serum (BDS) were used as cholesterol acceptors. The ILM method showed sufficient basic performance and a good correlation with the ILG method. Furthermore, when the CEC of 15 serum samples from healthy subjects was measured, a good correlation between HDL-cholesterol level and the ILG method was confirmed. Thus, it was confirmed that the ILM method was successfully developed and could be automated.
Collapse
|
35
|
Akash MSH, Rasheed S, Rehman K, Ibrahim M, Imran M, Assiri MA. Biochemical Activation and Regulatory Functions of Trans-Regulatory KLF14 and Its Association with Genetic Polymorphisms. Metabolites 2023; 13:metabo13020199. [PMID: 36837818 PMCID: PMC9962810 DOI: 10.3390/metabo13020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Krüpple-Like family of transcription factor-14 (KLF14) is a master trans-regulatory gene that has multiple biological regulatory functions and is involved in many pathological mechanisms. It controls the expressions of several other genes which are involved in multiple regulatory functions. KLF14 plays a significant role in lipid metabolism, glucose regulation and insulin sensitivity. Cell apoptosis, proliferation, and differentiation are regulated by the KLF14 gene, and up-regulation of KLF14 prevents cancer progression. KLF14 has been used as an epigenetic biomarker for the estimation of chronological age due to the presence of different age-related CpG sites on genes that become methylated with age. Different genome-wide association studies have identified several KLF14 variants in adipose tissues. These single nucleotide polymorphisms in KLF14 have been associated with dyslipidemia, insulin resistance, and glucose intolerance. Moreover, the prevalence of genetic polymorphism is different in different populations due to ethnic differences and epigenetic modifications. In addition, environmental and physiological factors such as diet, age, gender, and obesity are also responsible for genetic mutations in KLF14.
Collapse
Affiliation(s)
- Muhammad Sajid Hamid Akash
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad 38000, Pakistan
- Correspondence: (M.S.H.A.); (K.R.)
| | - Sumbal Rasheed
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, The Women University, Multan 60000, Pakistan
- Correspondence: (M.S.H.A.); (K.R.)
| | - Muhammad Ibrahim
- Department of Applied Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Imran
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 62413, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, Abha 62413, Saudi Arabia
| | - Mohammed A. Assiri
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 62413, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, Abha 62413, Saudi Arabia
| |
Collapse
|
36
|
Zhang C, Wu X, Shi P, Ma H, Fang F, Feng Q, Zhao S, Zhang R, Huang J, Xu X, Xiao W, Cao G, Ji X. Diterpenoids inhibit ox-LDL-induced foam cell formation in RAW264.7 cells by promoting ABCA1 mediated cholesterol efflux. Front Pharmacol 2023; 14:1066758. [PMID: 36713845 PMCID: PMC9877220 DOI: 10.3389/fphar.2023.1066758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Introduction: Atherosclerosis is the main cause of many cardiovascular diseases and contributes to morbidity and mortality worldwide. The formation of macrophage-derived foam cells plays a critical role in the early stage of atherosclerosis pathogenesis. Diterpenoids found in the flowers of Callicarpa rubella Lindl., a traditional Chinese medicine, have been reported to have anti-inflammatory activity. However, little is known about the effects of these diterpenoids on macrophage foam cell formation. Methods: A macrophage-derived foam cell formation model was established by treating RAW264.7 cells with oxidized low-density lipoprotein (ox-LDL) for 24 h. Oil red O staining were used to detect the intracellular lipids. The cholesterol efflux capacity was assayed by labeling cells with 22-NBD-cholesterol. Western blots and real-time PCRs were performed to quantify protein and mRNA expressions. Results: Two diterpenoid molecules, 14α-hydroxyisopimaric acid (C069002) and isopimaric acid (C069004), extracted from the flowers of Callicarpa rubella Lindl., significantly attenuated ox-LDL-induced foam cell formation in RAW264.7 macrophages. Further investigation showed that these two diterpenoids could promote cholesterol efflux from RAW264.7 macrophages to apolipoprotein A-I or high-density lipoproteins, which was associated with upregulated expression of ATP-binding cassette A1/G1 (ABCA1/G1), liver X receptor-α (LXRα), and peroxisome proliferator-activated receptor-γ (PPARγ). Unexpectedly, the diterpenoids C069002 and C069004 failed to enhance the mRNA transcription of the ABCG1 gene in macrophage-derived foam cells induced by ox-LDL. To evaluate the effects of diterpenoids on macrophage foam cell formation and determine the underlying mechanism, two drugs (lovastatin and rosiglitazone) were used as positive controls. Although both drugs could reduce macrophage foam cell formation and promote cholesterol efflux, they each had distinctive abilities to modulate the expression of cholesterol efflux-related genes. In contrast to lovastatin, rosiglitazone showed a similar influence on the expression of cholesterol efflux-related genes (including ABCA1, LXRα, and PPARγ) as the diterpenoids regardless of the presence or absence of ox-LDL, implying a similar mechanism by which they may exert atheroprotective effects. Conclusion: Our research indicates that diterpenoids effectively inhibit ox-LDL-induced macrophage foam cell formation by promoting cholesterol efflux from macrophages via the PPARγ-LXRα-ABCA1 pathway. Further investigation of diterpenoids as potential drugs for the treatment of atherosclerosis is warranted.
Collapse
Affiliation(s)
- Cheng Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Xuewen Wu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Pengmin Shi
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Hongyu Ma
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Fei Fang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Qianlang Feng
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Shuang Zhao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Ruipu Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Jinyuan Huang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China
| | - Xinting Xu
- Department of Pulmonary and Critical Care Medicine, Xi’an International Medical Center Hospital, Xi’an, China,*Correspondence: Xinting Xu, ; Weilie Xiao, ; Guang Cao, ; Xu Ji,
| | - Weilie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China,*Correspondence: Xinting Xu, ; Weilie Xiao, ; Guang Cao, ; Xu Ji,
| | - Guang Cao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China,*Correspondence: Xinting Xu, ; Weilie Xiao, ; Guang Cao, ; Xu Ji,
| | - Xu Ji
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Provincial Center for Research and Development of Natural Products, Yunnan Characteristic Plant Extraction Laboratory, Ministry of Education, School of Pharmacy, Yunnan University, Kunming, China,*Correspondence: Xinting Xu, ; Weilie Xiao, ; Guang Cao, ; Xu Ji,
| |
Collapse
|
37
|
HDL Functions-Current Status and Future Perspectives. Biomolecules 2023; 13:biom13010105. [PMID: 36671490 PMCID: PMC9855960 DOI: 10.3390/biom13010105] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in Western countries. A low HDL-C is associated with the development of CVD. However, recent epidemiology studies have shown U-shaped curves between HDL-C and CVD mortality, with paradoxically increased CVD mortality in patients with extremely high HDL-C levels. Furthermore, HDL-C raising therapy using nicotinic acids or CETP inhibitors mostly failed to reduce CVD events. Based on this background, HDL functions rather than HDL-C could be a novel biomarker; research on the clinical utility of HDL functionality is ongoing. In this review, we summarize the current status of HDL functions and their future perspectives from the findings of basic research and clinical trials.
Collapse
|
38
|
Qin Y, Medina MW. Mechanism of the Regulation of Plasma Cholesterol Levels by PI(4,5)P 2. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:89-119. [PMID: 36988878 DOI: 10.1007/978-3-031-21547-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Elevated low-density lipoprotein (LDL) cholesterol (LDLc) is one of the most well-established risk factors for cardiovascular disease, while high levels of high-density lipoprotein (HDL) cholesterol (HDLc) have been associated with protection from cardiovascular disease. Cardiovascular disease remains one of the leading causes of death worldwide; thus it is important to understand mechanisms that impact LDLc and HDLc metabolism. In this chapter, we will discuss molecular processes by which phosphatidylinositol-(4,5)-bisphosphate, PI(4,5)P2, is thought to modulate LDLc or HDLc. Section 1 will provide an overview of cholesterol in the circulation, discussing processes that modulate the various forms of lipoproteins (LDL and HDL) carrying cholesterol. Section 2 will describe how a PI(4,5)P2 phosphatase, transmembrane protein 55B (TMEM55B), impacts circulating LDLc levels through its ability to regulate lysosomal decay of the low-density lipoprotein receptor (LDLR), the primary receptor for hepatic LDL uptake. Section 3 will discuss how PI(4,5)P2 interacts with apolipoprotein A-I (apoA1), the key apolipoprotein on HDL. In addition to direct mechanisms of PI(4,5)P2 action on circulating cholesterol, Sect. 4 will review how PI(4,5)P2 may indirectly impact LDLc and HDLc by affecting insulin action. Last, as cholesterol is controlled through intricate negative feedback loops, Sect. 5 will describe how PI(4,5)P2 is regulated by cholesterol.
Collapse
Affiliation(s)
- Yuanyuan Qin
- Department of Pediatrics, Division of Cardiology, University of California, San Francisco, Oakland, CA, USA
| | - Marisa W Medina
- Department of Pediatrics, Division of Cardiology, University of California, San Francisco, Oakland, CA, USA.
| |
Collapse
|
39
|
Zhang W, Li W, Tian R, Cao L. High-density lipoprotein level is associated with hemorrhage transformation after ischemic stroke treatment with intravenous thrombolysis: A systematic review and meta-analysis. J Clin Neurosci 2022; 106:122-127. [DOI: 10.1016/j.jocn.2022.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/15/2022]
|
40
|
Wu Z, Li X, Wen Q, Tao B, Qiu B, Zhang Q, Wang J. Serum LDL-C/HDL-C ratio and the risk of carotid plaques: a longitudinal study. BMC Cardiovasc Disord 2022; 22:501. [PMID: 36434516 PMCID: PMC9700971 DOI: 10.1186/s12872-022-02942-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Dyslipidemia contributes to an increased risk of carotid atherosclerosis. However, the association between the ratio of low-density lipoprotein cholesterol (LDL-C) to high-density lipoprotein cholesterol (HDL-C) and carotid plaque formation has not been well documented. This study aims to assess the role of LDL-C/HDL-C in the risk of carotid plaque formation in a Chinese population. METHODS We followed 2,191 participants who attended the annual routine health examination. Cox proportional hazards regression, restricted cubic spline (RCS), and subgroup analysis were applied to evaluate the association between the LDL-C/HDL-C ratio and carotid plaques. The hazard ratio (HR) and 95% confidence interval (CI) were used to estimate the strength of the association. RESULTS Among 2,191 participants, 388 had incident carotid plaques detected, with a median follow-up time of 1.05 years. Compared with subjects younger than 45 years, those aged 45 to 59 years (HR: 2.00, 95% CI: 1.55-2.58) and over 60 years (HR: 3.36, 95% CI: 2.47-4.58) had an increased risk of carotid plaque formation. Males (HR: 1.26, 95% CI: 1.01-1.56), diabetes (HR: 1.46, 95% CI: 1.06-2.01) and a high LDL-C/HDL-C ratio (HR: 1.22, 95% CI: 1.07-1.38) were significantly linked with the occurrence of carotid plaques. After adjusting for potential confounding factors, we observed that a high LDL-C/HDL-C ratio promoted carotid plaque events (HR: 1.30, 95% CI: 1.12-1.50). The RCS analysis revealed a significant nonlinear association. The association was stronger among females (P-interaction < 0.05). CONCLUSION A high LDL-C/HDL-C ratio could accelerate the occurrence of carotid plaques. Older men with diabetes and dyslipidemia are the critical target population. Women may be more likely to benefit from lipid-lowering interventions and thus avoid carotid plaque formation.
Collapse
Affiliation(s)
- Zhuchao Wu
- grid.89957.3a0000 0000 9255 8984Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Xiaona Li
- grid.412676.00000 0004 1799 0784Department of Health Management Center, the First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, China ,grid.89957.3a0000 0000 9255 8984Department of Health Management, Center for Global Health, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Qin Wen
- grid.89957.3a0000 0000 9255 8984Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Bilin Tao
- grid.89957.3a0000 0000 9255 8984Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Beibei Qiu
- grid.89957.3a0000 0000 9255 8984Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Qun Zhang
- grid.412676.00000 0004 1799 0784Department of Health Management Center, the First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, China ,grid.89957.3a0000 0000 9255 8984Department of Health Management, Center for Global Health, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| | - Jianming Wang
- grid.89957.3a0000 0000 9255 8984Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, 211166 Nanjing, China
| |
Collapse
|
41
|
Brodeur MR, Rhainds D, Charpentier D, Boulé M, Mihalache-Avram T, Mecteau M, Brand G, Pedneault-Gagnon V, Fortier A, Niesor EJ, Rhéaume E, Maugeais C, Tardif JC. Dalcetrapib and anacetrapib increase apolipoprotein E-containing HDL in rabbits and humans. J Lipid Res 2022; 64:100316. [PMID: 36410424 PMCID: PMC9793321 DOI: 10.1016/j.jlr.2022.100316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 10/20/2022] [Accepted: 11/11/2022] [Indexed: 11/20/2022] Open
Abstract
The large HDL particles generated by administration of cholesteryl ester transfer protein inhibitors (CETPi) remain poorly characterized, despite their potential importance in the routing of cholesterol to the liver for excretion, which is the last step of the reverse cholesterol transport. Thus, the effects of the CETPi dalcetrapib and anacetrapib on HDL particle composition were studied in rabbits and humans. The association of rabbit HDL to the LDL receptor (LDLr) in vitro was also evaluated. New Zealand White rabbits receiving atorvastatin were treated with dalcetrapib or anacetrapib. A subset of patients from the dal-PLAQUE-2 study treated with dalcetrapib or placebo were also studied. In rabbits, dalcetrapib and anacetrapib increased HDL-C by more than 58% (P < 0.01) and in turn raised large apo E-containing HDL by 66% (P < 0.001) and 59% (P < 0.01), respectively. Additionally, HDL from CETPi-treated rabbits competed with human LDL for binding to the LDLr on HepG2 cells more than control HDL (P < 0.01). In humans, dalcetrapib increased concentrations of large HDL particles (+69%, P < 0.001) and apo B-depleted plasma apo E (+24%, P < 0.001), leading to the formation of apo E-containing HDL (+47%, P < 0.001) devoid of apo A-I. Overall, in rabbits and humans, CETPi increased large apo E-containing HDL particle concentration, which can interact with hepatic LDLr. The catabolism of these particles may depend on an adequate level of LDLr to contribute to reverse cholesterol transport.
Collapse
Affiliation(s)
| | | | | | - Marie Boulé
- Montreal Heart Institute, Montreal, Quebec, Canada
| | | | | | | | | | - Annik Fortier
- Montreal Health Innovations Coordinating Center, Montreal, Quebec, Canada
| | | | - Eric Rhéaume
- Montreal Heart Institute, Montreal, Quebec, Canada,Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | | | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec, Canada; Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
42
|
Yan L, Guo J, Gao X. Expression of SR-BI and LXR α in Macrophage Origin Foam Cells of Mouse RAW264.7 Up-regulated by Astragalus Polysaccharide. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:3920584. [PMID: 36247857 PMCID: PMC9532143 DOI: 10.1155/2022/3920584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/07/2022] [Accepted: 09/16/2022] [Indexed: 01/26/2023]
Abstract
Objective To investigate the influences of Astragalus polysaccharides (APS) on the expressions of SR-BI(Scavenger receptor-BI) and LXR α in RAW264.7 macrophage origin foam cells. Methods Mouse RAW264.7 cells were induced in foam cells and identification of the foamed by oil-red O staining. The RAW264.7 macrophage origin foam cells were dealt with APS with distinct contents (0, 10, 20, 50, or 100 mg/L). The mRNA and protein expressions of SR-BI and LXRα were measured by RT-PCR as well as ELISA, respectively. Results Macrophages were differentiated into foam cells 48 h after ox-LDL induction. In contrast to the control part, the mRNA and protein expressions of SR-BI and LXRα in RAW264.7 macrophage origin foam cells were up-regulated dosage-dependently after being treated with different concentrations of APS (P < 0.05). Conclusion APS could promote intracellular cholesterol efflux by up-regulating the expressions of SR-BI and LXR α of RAW264.7 cells.
Collapse
Affiliation(s)
- Lichen Yan
- Department of Gastroenterology, Jincheng People's Hospital of Shanxi, Jincheng 048000, Shanxi, China
| | - Jing Guo
- Heping Hospital Affiliated to Changzhi Medical College, Changzhi 046000, Shanxi, China
| | - Xiaohua Gao
- Heping Hospital Affiliated to Changzhi Medical College, Changzhi 046000, Shanxi, China
| |
Collapse
|
43
|
Bonaca MP, Morrow DA, Bergmark BA, Berg DD, Lima JAC, Hoffmann U, Kato Y, Lu MT, Kuder J, Murphy SA, Spinar J, Oude Ophuis T, Kiss RG, Lopez-Sendon J, Averkov O, Wheatcroft SB, Kubica J, Carlos Nicolau J, Furtado RHM, Abuhatzira L, Hirshberg B, Omar SA, Vavere AL, Chang YT, George RT, Sabatine MS. Randomized, Placebo-Controlled Phase 2b Study to Evaluate the Safety and Efficacy of Recombinant Human Lecithin Cholesterol Acyltransferase in Acute ST-Segment-Elevation Myocardial Infarction: Results of REAL-TIMI 63B. Circulation 2022; 146:907-916. [PMID: 36039762 DOI: 10.1161/circulationaha.122.059325] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND High-density lipoprotein plays a key role in reverse cholesterol transport. In addition, high-density lipoprotein particles may be cardioprotective and reduce infarct size in the setting of myocardial injury. Lecithin-cholesterol acyltransferase is a rate-limiting enzyme in reverse cholesterol transport. MEDI6012 is a recombinant human lecithin-cholesterol acyltransferase that increases high-density lipoprotein cholesterol. Administration of lecithin-cholesterol acyltransferase has the potential to reduce infarct size and regress coronary plaque in acute ST-segment-elevation myocardial infarction. METHODS REAL-TIMI 63B (A Randomized, Placebo‑controlled Phase 2b Study to Evaluate the Safety and Efficacy of MEDI6012 in Acute ST Elevation Myocardial Infarction) was a phase 2B multinational, placebo-controlled, randomized trial. Patients with ST-segment-elevation myocardial infarction within 6 hours of symptom onset and planned for percutaneous intervention were randomly assigned 2:1 to MEDI6012 (2- or 6-dose regimen) or placebo and followed for 12 weeks. The primary outcome was infarct size as a percentage of left ventricular mass by cardiac MRI at 10 to 12 weeks, with the primary analysis in patients with TIMI Flow Grade 0 to 1 before percutaneous intervention who received at least 2 doses of MEDI6012. The secondary outcome was change in noncalcified plaque volume on coronary computed tomographic angiography from baseline to 10 to 12 weeks with the primary analysis in patients who received all 6 doses of MEDI6012. RESULTS A total of 593 patients were randomly assigned. Patients were a median of 62 years old, 77.9% male, and 95.8% statin naive. Median time from symptom onset to randomization was 146 (interquartile range [IQR], 103-221) minutes and from hospitalization to randomization was 12.7 (IQR, 6.6-24.0) minutes, and the first dose of drug was administered a median of 8 (IQR, 3-13) minutes before percutaneous intervention. The index myocardial infarction was anterior in 69.6% and TIMI Flow Grade 0 to 1 in 65.1% of patients. At 12 weeks, infarct size did not differ between treatment groups (MEDI6012: 9.71%, IQR 4.79-16.38; placebo: 10.48%, [IQR, 4.92-16.61], 1-sided P=0.79. There was also no difference in noncalcified plaque volume (geometric mean ratio, 0.96 [95% CI, NA-1.10], 1-sided P=0.30). There was no significant difference in treatment emergent serious adverse events. CONCLUSIONS Administration of MEDI6012 in patients with acute ST-segment-elevation myocardial infarction did not result in a significant reduction in infarct size or noncalcified plaque volume at 12 weeks. MEDI6012 was well tolerated with no excess in overall serious adverse events. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03578809.
Collapse
Affiliation(s)
- Marc P Bonaca
- CPC Clinical Research, Department of Medicine, University of Colorado Anschutz School of Medicine, Aurora (M.P.B.)
| | - David A Morrow
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - Brian A Bergmark
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - David D Berg
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, MD (J.A.C.L., Y.K.)
| | - Udo Hoffmann
- Internal Cardioangiology Department, St. Ann University Hospital and Masaryk University, Brno, Czechia (J.S.)
| | - Yoko Kato
- Division of Cardiology, Johns Hopkins University, Baltimore, MD (J.A.C.L., Y.K.)
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston (U.H., M.T.L.)
| | - Julia Kuder
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - Sabina A Murphy
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| | - Jindrich Spinar
- Internal Cardioangiology Department, St. Ann University Hospital and Masaryk University, Brno, Czechia (J.S.)
| | - Ton Oude Ophuis
- Canisius Wilhelmina Ziekenhuis, Nijmegen, The Netherlands (T.O.O.)
| | - Róbert G Kiss
- Department of Cardiology, Military Hospital, Budapest, Hungary (R.G.K.)
| | - Jose Lopez-Sendon
- IdiPaz Research Institute, Hospital Universitario La Paz, UAM, Madrid, Spain (J.L.-S.)
| | - Oleg Averkov
- Pirogov Russian National Research Medical University, Moscow (O.A.)
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK (S.B.W.)
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland (J. Kubica)
| | - Jose Carlos Nicolau
- Instituto do Coracao, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Brazil (J.C.N., R.H.M.F.)
| | - Remo H M Furtado
- Instituto do Coracao, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Brazil (J.C.N., R.H.M.F.).,Academic Research Organization, Hospital Israelita Albert Einstein, Sao Paulo, Brazil (R.H.M.G.)
| | | | | | - Sami A Omar
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (S.A.O., A.L.V., Y.-T.C., R.T.G.)
| | - Andrea L Vavere
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (S.A.O., A.L.V., Y.-T.C., R.T.G.)
| | - Yi-Ting Chang
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (S.A.O., A.L.V., Y.-T.C., R.T.G.)
| | - Richard T George
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD (S.A.O., A.L.V., Y.-T.C., R.T.G.)
| | - Marc S Sabatine
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.A.M., B.A.B., D.D.B., J. Kuder, S.A.M., M.S.S.)
| |
Collapse
|
44
|
Hu HJ, Wang XH, Zhang TQ, Liu Y, Chen ZR, Zhang ZZ, Huang H, Tang HF, Jiang ZS. PLK1 promotes cholesterol efflux and alleviates atherosclerosis by up-regulating ABCA1 and ABCG1 expression via the AMPK/PPARγ/LXRα pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159221. [PMID: 35981705 DOI: 10.1016/j.bbalip.2022.159221] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 07/31/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase involving lipid metabolism and cardiovascular disease. However, its role in atherogenesis has yet to be determined. The aim of this study was to observe the impact of PLK1 on macrophage lipid accumulation and atherosclerosis development and to explore the underlying mechanisms. We found a significant reduction of PLK1 expression in lipid-loaded macrophages and atherosclerosis model mice. Lentivirus-mediated overexpression of PLK1 promoted cholesterol efflux and inhibited lipid accumulation in THP-1 macrophage-derived foam cells. Mechanistic analysis revealed that PLK1 stimulated the phosphorylation of AMP-activated protein kinase (AMPK), leading to activation of the peroxisome proliferator-activated receptor γ (PPARγ)/liver X receptor α (LXRα) pathway and up-regulation of ATP binding cassette transporter A1 (ABCA1) and ABCG1 expression. Injection of lentiviral vector expressing PLK1 increased reverse cholesterol transport, improved plasma lipid profiles and decreased atherosclerotic lesion area in apoE-deficient mice fed a Western diet. PLK1 overexpression also facilitated AMPK and HSL phosphorylation and enhanced the expression of PPARγ, LXRα, ABCA1, ABCG1 and LPL in the aorta. In summary, these data suggest that PLK1 inhibits macrophage lipid accumulation and mitigates atherosclerosis by promoting ABCA1- and ABCG1-dependent cholesterol efflux via the AMPK/PPARγ/LXRα pathway.
Collapse
Affiliation(s)
- Heng-Jing Hu
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of Cardiovascular Disease and Key Lab for Atherosclerosis of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiu-Heng Wang
- The First Affiliated Hospital, Department of Medical-record, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Tian-Qing Zhang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yao Liu
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zheng-Rong Chen
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhi-Zhu Zhang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hong Huang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hui-Fang Tang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhi-Sheng Jiang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of Cardiovascular Disease and Key Lab for Atherosclerosis of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
45
|
Abstract
The lymphatic vessels play an essential role in maintaining immune and fluid homeostasis and in the transport of dietary lipids. The discovery of lymphatic endothelial cell-specific markers facilitated the visualization and mechanistic analysis of lymphatic vessels over the past two decades. As a result, lymphatic vessels have emerged as a crucial player in the pathogenesis of several cardiovascular diseases, as demonstrated by worsened disease progression caused by perturbations to lymphatic function. In this review, we discuss the major findings on the role of lymphatic vessels in cardiovascular diseases such as hypertension, obesity, atherosclerosis, myocardial infarction, and heart failure.
Collapse
Affiliation(s)
- Dakshnapriya Balasubbramanian
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, Texas 77807, USA
| |
Collapse
|
46
|
HDL, cholesterol efflux, and ABCA1: Free from good and evil dualism. J Pharmacol Sci 2022; 150:81-89. [DOI: 10.1016/j.jphs.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/15/2022] [Accepted: 07/25/2022] [Indexed: 11/19/2022] Open
|
47
|
Independent Effects of Kidney Function and Cholesterol Efflux on Cardiovascular Mortality. Biomedicines 2022; 10:biomedicines10081832. [PMID: 36009383 PMCID: PMC9404976 DOI: 10.3390/biomedicines10081832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Impaired renal function is associated with cardiovascular and all-cause mortality. In the general population, HDL-cholesterol is associated with cardiovascular events, which is not true in patients with chronic kidney disease (CKD). This has been attributed to abnormal HDL function in CKD. Methods: In this study, we analyzed the association of genetic markers for kidney function with cholesterol efflux capacity as one of the major HDL functions, as well as with cardiovascular mortality, in 2469 patients of the Ludwigshafen Risk and Cardiovascular Health Study who all underwent coronary angiography. Results: A genetic score of 53 SNPs associated with GRF and the uromodulin SNP rs12917707 were inversely correlated with cholesterol efflux capacity. This was in line with the observed association between cholesterol efflux capacity and kidney function in these patients. Adjustment for eGFR and uromodulin as markers of kidney function did not affect the relationship between cholesterol efflux and cardiovascular mortality. Conclusions: Our data propose the view that cholesterol efflux and kidney function are exerting their effects on cardiovascular mortality via different and independent pathways. Decreased cholesterol efflux may therefore not mediate the effects of impaired kidney function on cardiovascular mortality.
Collapse
|
48
|
Zou J, Xu C, Zhao ZW, Yin SH, Wang G. Asprosin inhibits macrophage lipid accumulation and reduces atherosclerotic burden by up-regulating ABCA1 and ABCG1 expression via the p38/Elk-1 pathway. Lab Invest 2022; 20:337. [PMID: 35902881 PMCID: PMC9331044 DOI: 10.1186/s12967-022-03542-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/17/2022] [Indexed: 12/27/2022]
Abstract
Background Asprosin, a newly discovered adipokine, is a C-terminal cleavage product of profibrillin. Asprosin has been reported to participate in lipid metabolism and cardiovascular disease, but its role in atherogenesis remains elusive. Methods Asprosin was overexpressed in THP-1 macrophage-derived foam cells and apoE−/− mice using the lentiviral vector. The expression of relevant molecules was determined by qRT-PCR and/or western blot. The intracellular lipid accumulation was evaluated by high-performance liquid chromatography and Oil red O staining. HE and Oil red O staining was employed to assess plaque burden in vivo. Reverse cholesterol transport (RCT) efficiency was measured using [3H]-labeled cholesterol. Results Exposure of THP-1 macrophages to oxidized low-density lipoprotein down-regulated asprosin expression. Lentivirus-mediated overexpression of asprosin promoted cholesterol efflux and inhibited lipid accumulation in THP-1 macrophage-derived foam cells. Mechanistic analysis revealed that asprosin overexpression activated p38 and stimulated the phosphorylation of ETS-like transcription factor (Elk-1) at Ser383, leading to Elk-1 nuclear translocation and the transcriptional activation of ATP binding cassette transporters A1 (ABCA1) and ABCG1. Injection of lentiviral vector expressing asprosin diminished atherosclerotic lesion area, increased plaque stability, improved plasma lipid profiles and facilitated RCT in apoE−/− mice. Asprosin overexpression also increased the phosphorylation of p38 and Elk-1 as well as up-regulated the expression of ABCA1 and ABCG1 in the aortas. Conclusion Asprosin inhibits lipid accumulation in macrophages and decreases atherosclerotic burden in apoE−/− mice by up-regulating ABCA1 and ABCG1 expression via activation of the p38/Elk-1 signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03542-0.
Collapse
Affiliation(s)
- Jin Zou
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Can Xu
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Shan-Hui Yin
- The First Affiliated Hospital, Department of Neonatology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Gang Wang
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
49
|
Effect of N-3 Polyunsaturated Fatty Acids on Lipid Composition in Familial Hypercholesterolemia: A Randomized Crossover Trial. Biomedicines 2022; 10:biomedicines10081809. [PMID: 36009356 PMCID: PMC9405021 DOI: 10.3390/biomedicines10081809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Individuals with familial hypercholesterolemia (FH) have an increased risk of cardiovascular disease. Treatment is mainly low-density lipoprotein cholesterol (LDL-C) reduction. How omega-3 polyunsaturated fatty acids (n-3 PUFAs) supplements affect lipoproteins in FH subjects is unknown. We hypothesized that a high-dose n-3 PUFA supplement would reduce atherogenic lipoproteins and influence the high-density lipoprotein cholesterol (HDL-C) function. We performed a randomized, double-blinded crossover study with 34 genetically verified FH individuals (18−75 years, clinically stable, statin treatment > 12 months). Treatment was 4 g n-3 PUFAs (1840 mg eicosapentaenoic acid and 1520 mg docosahexaenoic acid daily) or four capsules of olive oil for three months in a crossover design with a washout period of three months. The defined outcomes were changes in triglycerides, lipoproteins, lipoprotein subfractions, apolipoproteins, and HDL-C function. After treatment with n-3 PUFAs, total cholesterol, LDL-C, and triglycerides were reduced compared to placebo (p ≤ 0.01 for all). Total HDL-C levels were unchanged, but the subfraction of large HDL-C was higher (p ≤ 0.0001) after n-3 PUFAs than after placebo, and intermediate HDL-C and small HDL-C were reduced after n-3 PUFAs compared to placebo (p = 0.02 and p ≤ 0.001, respectively). No changes were found in apolipoproteins and HDL-C function. N-3 PUFAs supplements reduced atherogenic lipoproteins in FH subjects, leaving HDL-C function unaffected.
Collapse
|
50
|
Chen L, Zhao ZW, Zeng PH, Zhou YJ, Yin WJ. Molecular mechanisms for ABCA1-mediated cholesterol efflux. Cell Cycle 2022; 21:1121-1139. [PMID: 35192423 PMCID: PMC9103275 DOI: 10.1080/15384101.2022.2042777] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The maintenance of cellular cholesterol homeostasis is essential for normal cell function and viability. Excessive cholesterol accumulation is detrimental to cells and serves as the molecular basis of many diseases, such as atherosclerosis, Alzheimer's disease, and diabetes mellitus. The peripheral cells do not have the ability to degrade cholesterol. Cholesterol efflux is therefore the only pathway to eliminate excessive cholesterol from these cells. This process is predominantly mediated by ATP-binding cassette transporter A1 (ABCA1), an integral membrane protein. ABCA1 is known to transfer intracellular free cholesterol and phospholipids to apolipoprotein A-I (apoA-I) for generating nascent high-density lipoprotein (nHDL) particles. nHDL can accept more free cholesterol from peripheral cells. Free cholesterol is then converted to cholesteryl ester by lecithin:cholesterol acyltransferase to form mature HDL. HDL-bound cholesterol enters the liver for biliary secretion and fecal excretion. Although how cholesterol is transported by ABCA1 to apoA-I remains incompletely understood, nine models have been proposed to explain this effect. In this review, we focus on the current view of the mechanisms underlying ABCA1-mediated cholesterol efflux to provide an important framework for future investigation and lipid-lowering therapy.
Collapse
Affiliation(s)
- Lei Chen
- Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Peng-Hui Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ying-Jie Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wen-Jun Yin
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China,CONTACT Wen-Jun Yin Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan421001, China
| |
Collapse
|