1
|
Xu M, Wang W, Cheng J, Qu H, Xu M, Wang L. Effects of mitochondrial dysfunction on cellular function: Role in atherosclerosis. Biomed Pharmacother 2024; 174:116587. [PMID: 38636397 DOI: 10.1016/j.biopha.2024.116587] [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: 12/20/2023] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Atherosclerosis, an immunoinflammatory disease of medium and large arteries, is associated with life-threatening clinical events, such as acute coronary syndromes and stroke. Chronic inflammation and impaired lipoprotein metabolism are considered to be among the leading causes of atherosclerosis, while numerous risk factors, including arterial hypertension, diabetes mellitus, obesity, and aging, can contribute to the development of the disease. In recent years, emerging evidence has underlined the key role of mitochondrial dysfunction in the pathogenesis of atherosclerosis. Mitochondrial dysfunction is believed to result in an increase in reactive oxygen species, leading to oxidative stress, chronic inflammation, and intracellular lipid deposition, all of which can contribute to the pathogenesis of atherosclerosis. Critical cells, including endothelial cells, vascular smooth muscle cells, and macrophages, play an important role in atherosclerosis. Mitochondrial function is also involved in maintaining the normal function of these cells. To better understand the relationship between mitochondrial dysfunction and atherosclerosis, this review summarizes the findings of recent studies and discusses the role of mitochondrial dysfunction in the risk factors and critical cells of atherosclerosis. FACTS: OPEN QUESTIONS.
Collapse
Affiliation(s)
- Minwen Xu
- Clinical Skills Center, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Wenjun Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jingpei Cheng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; Basic Medical College, Gannan Medical University, Ganzhou 341000, China
| | - Hongen Qu
- Gannan Normal University, Ganzhou 341000, China.
| | - Minjuan Xu
- Department of Obstetrics and Gynecology, Ganzhou People's Hospital, Ganzhou 341000, China.
| | - Liefeng Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; Basic Medical College, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
2
|
Zhang Y, Luo S, Gao Y, Tong W, Sun S. High-Density Lipoprotein Subfractions Remodeling: A Critical Process for the Treatment of Atherosclerotic Cardiovascular Diseases. Angiology 2024; 75:441-453. [PMID: 36788038 DOI: 10.1177/00033197231157473] [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] [Indexed: 02/16/2023]
Abstract
Numerous studies have shown that a low level of high-density lipoprotein cholesterol (HDL-C) is an independent biomarker of cardiovascular disease. High-density lipoprotein (HDL) is considered to be a protective factor for atherosclerosis (AS). Therefore, raising HDL-C has been widely recognized as a promising strategy to treat atherosclerotic cardiovascular diseases (ASCVD). However, several studies have found that increasing HDL-C levels does not necessarily reduce the risk of ASCVD. HDL particles are highly heterogeneous in structure, composition, and biological function. Moreover, HDL particles from atherosclerotic patients exhibit impaired anti-atherogenic functions and these dysfunctional HDL particles might even promote ASCVD. This makes it uncertain that HDL-raising therapy will prevent and treat ASCVD. It is necessary to comprehensively analyze the structure and function of HDL subfractions. We review current advances related to HDL subfractions remodeling and highlight how current lipid-modifying drugs such as niacin, statins, fibrates, and cholesteryl ester transfer protein inhibitors regulate cholesterol concentration of HDL and specific HDL subfractions.
Collapse
Affiliation(s)
- Yaling Zhang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Shiyu Luo
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Yi Gao
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, China
| | - Shaowei Sun
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
3
|
Zhou SM, Lin FM, Mu CK, Wang CL, Zhou QC, Sun P, Yin F. Cellular localization and potential ligands of a novel scavenger receptor class B/CD36 protein homolog (Pt-SRB2) identified in the marine crab, Portunustrituberculatus. FISH & SHELLFISH IMMUNOLOGY 2024; 145:109355. [PMID: 38168634 DOI: 10.1016/j.fsi.2023.109355] [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: 10/19/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
The scavenger receptor class B family proteins (SRB) are multiligand membrane receptor proteins. Herein, a novel SRB homolog (Pt-SRB2) was identified in Portunus trituberculatus. The open reading frame of Pt-SRB2 was predicted to encode 520 amino acid residues comprising a typical CD36 domain. Phylogenetic analysis showed that Pt-SRB2 distinctly clustered with the SRB homologs of most crustaceans and Drosophila but was separate from all vertebrate CD36/SRB. Semi-quantitative and Real-time quantitative PCR revealed that the abundance of Pt-SRB2 transcripts was the highest in hepatopancreas than in other tested tissues. Overexpressed Pt-SRB2 was distributed primarily in the cell membrane and cytoplasm of HEK293T or Drosophila Schneider 2 cells. In crab hemocytes, Pt-SRB2 was distributed primarily in the cell membrane by immunofluorescence staining. In addition, the immunofluorescence staining showed that green fluorescence signals were mainly located in the inner lumen membrane of the hepatopancreatic tubules. Moreover, solid-phase enzyme-linked immunosorbent assay revealed that rPt-SRB2-L exhibited relative high affinity with lipopolysaccharides, and relative moderate binding affinity with lipoteichoic acid or peptidoglycan. Of note, rPt-SRB2-L showed high binding affinity with eicosapentaenoic acid among a series of long-chain polyunsaturated fatty acids. Taken together, this study provided valuable data for understanding the functions of the crab CD36/SRB.
Collapse
Affiliation(s)
- Su-Ming Zhou
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Fang-Mei Lin
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Chang-Kao Mu
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Chun-Lin Wang
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Qi-Cun Zhou
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Peng Sun
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Fei Yin
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
4
|
Asoudeh M, Nguyen N, Raith M, Denman DS, Anozie UC, Mokhtarnejad M, Khomami B, Skotty KM, Isaac S, Gebhart T, Vaigneur L, Gelgie A, Dego OK, Freeman T, Beever J, Dalhaimer P. PEGylated nanoparticles interact with macrophages independently of immune response factors and trigger a non-phagocytic, low-inflammatory response. J Control Release 2024; 366:282-296. [PMID: 38123071 PMCID: PMC10922886 DOI: 10.1016/j.jconrel.2023.12.019] [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: 09/01/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Poly-ethylene-glycol (PEG)-based nanoparticles (NPs) - including cylindrical micelles (CNPs), spherical micelles (SNPs), and PEGylated liposomes (PLs) - are hypothesized to be cleared in vivo by opsonization followed by liver macrophage phagocytosis. This hypothesis has been used to explain the rapid and significant localization of NPs to the liver after administration into the mammalian vasculature. Here, we show that the opsonization-phagocytosis nexus is not the major factor driving PEG-NP - macrophage interactions. First, mouse and human blood proteins had insignificant affinity for PEG-NPs. Second, PEG-NPs bound macrophages in the absence of serum proteins. Third, lipoproteins blocked PEG-NP binding to macrophages. Because of these findings, we tested the postulate that PEG-NPs bind (apo)lipoprotein receptors. Indeed, PEG-NPs triggered an in vitro macrophage transcription program that was similar to that triggered by lipoproteins and different from that triggered by lipopolysaccharide (LPS) and group A Streptococcus. Unlike LPS and pathogens, PLs did not increase transcripts involved in phagocytosis or inflammation. High-density lipoprotein (HDL) and SNPs triggered remarkably similar mouse bone-marrow-derived macrophage transcription programs. Unlike opsonized pathogens, CNPs, SNPs, and PLs lowered macrophage autophagosome levels and either reduced or did not increase the secretion of key macrophage pro-inflammatory cytokines and chemokines. Thus, the sequential opsonization and phagocytosis process is likely a minor aspect of PEG-NP - macrophage interactions. Instead, PEG-NP interactions with (apo)lipoprotein and scavenger receptors appear to be a strong driving force for PEG-NP - macrophage binding, entry, and downstream effects. We hypothesize that the high presence of these receptors on liver macrophages and on liver sinusoidal endothelial cells is the reason PEG-NPs localize rapidly and strongly to the liver.
Collapse
Affiliation(s)
- Monireh Asoudeh
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Nicole Nguyen
- School of Medical Laboratory Science, University of Tennessee Medical Center, Knoxville, TN 37996, USA
| | - Mitch Raith
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Desiree S Denman
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Uche C Anozie
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Mahshid Mokhtarnejad
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Bamin Khomami
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Kaitlyn M Skotty
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Sami Isaac
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | - Aga Gelgie
- Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | | | - Trevor Freeman
- Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Jon Beever
- Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Paul Dalhaimer
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA.
| |
Collapse
|
5
|
Feng N, Feng Y, Tan J, Zhou C, Xu J, Chen Y, Xiao J, He Y, Wang C, Zhou M, Wu Q. Inhibition of advance glycation end products formation, gastrointestinal digestion, absorption and toxicity: A comprehensive review. Int J Biol Macromol 2023; 249:125814. [PMID: 37451379 DOI: 10.1016/j.ijbiomac.2023.125814] [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: 01/17/2023] [Revised: 06/18/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Advanced glycation end-products (AGEs) are the final products of the non-enzymatic interaction between reducing sugars and amino groups in proteins, lipids and nucleic acids. In numerous diseases, such as diabetes, neuropathy, atherosclerosis, aging, nephropathy, retinopathy, and chronic renal illness, accumulation of AGEs has been proposed as a pathogenic mechanism of inflammation, oxidative stress, and structural tissue damage leading to chronic vascular issues. Current studies on the inhibition of AGEs mainly focused on food processing. However, there are few studies on the inhibition of AGEs during digestion, absorption and metabolism although there are still plenty of AGEs in our body with our daily diet. This review comprehensively expounded AGEs inhibition mechanism based on the whole process of digestion, absorption and metabolism by polyphenols, amino acids, hydrophilic colloid, carnosine and other new anti-glycation agents. Our study will provide a ground-breaking perspective on mediation or inhibition AGEs.
Collapse
Affiliation(s)
- Nianjie Feng
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Yingna Feng
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Jiangying Tan
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Chen Zhou
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Jianhua Xu
- Pinyuan (Suizhou) Modern Agriculture Development Co., LTD., Suizhou, Hubei 441300, China
| | - Yashu Chen
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan, China
| | - Juan Xiao
- State Key Laboratory of Marine Resource Utilization in South China Sea, Ministry of Education, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Engineering Research Center of Utilization of Tropical Polysaccharide Resources, School of Food Science and Engineering, Hainan University, Haikou, China
| | - Yi He
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chao Wang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Mengzhou Zhou
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Qian Wu
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| |
Collapse
|
6
|
Pang J, Raka F, Heirali AA, Shao W, Liu D, Gu J, Feng JN, Mineo C, Shaul PW, Qian X, Coburn B, Adeli K, Ling W, Jin T. Resveratrol intervention attenuates chylomicron secretion via repressing intestinal FXR-induced expression of scavenger receptor SR-B1. Nat Commun 2023; 14:2656. [PMID: 37160898 PMCID: PMC10169763 DOI: 10.1038/s41467-023-38259-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 04/21/2023] [Indexed: 05/11/2023] Open
Abstract
Two common features of dietary polyphenols have hampered our mechanistic understanding of their beneficial effects for decades: targeting multiple organs and extremely low bioavailability. We show here that resveratrol intervention (REV-I) in high-fat diet (HFD)-challenged male mice inhibits chylomicron secretion, associated with reduced expression of jejunal but not hepatic scavenger receptor class B type 1 (SR-B1). Intestinal mucosa-specific SR-B1-/- mice on HFD-challenge exhibit improved lipid homeostasis but show virtually no further response to REV-I. SR-B1 expression in Caco-2 cells cannot be repressed by pure resveratrol compound while fecal-microbiota transplantation from mice on REV-I suppresses jejunal SR-B1 in recipient mice. REV-I reduces fecal levels of bile acids and activity of fecal bile-salt hydrolase. In Caco-2 cells, chenodeoxycholic acid treatment stimulates both FXR and SR-B1. We conclude that gut microbiome is the primary target of REV-I, and REV-I improves lipid homeostasis at least partially via attenuating FXR-stimulated gut SR-B1 elevation.
Collapse
Affiliation(s)
- Juan Pang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, PR China
- Division of Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital, Sichuan University, Chengdu, PR China
| | - Fitore Raka
- Department of Molecular Structure and Function Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Alya Abbas Heirali
- Department of Medicine, Division of Infectious Diseases, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Weijuan Shao
- Division of Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Dinghui Liu
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Jianqiu Gu
- Department of Endocrinology and Metabolism and The Institute of Endocrinology, The First Hospital of China Medical University, Shenyang, PR China
| | - Jia Nuo Feng
- Division of Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Chieko Mineo
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philip W Shaul
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoxian Qian
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Bryan Coburn
- Department of Medicine, Division of Infectious Diseases, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Khosrow Adeli
- Department of Molecular Structure and Function Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, PR China.
| | - Tianru Jin
- Division of Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
7
|
Wang YE, Kirschke CP, Woodhouse LR, Bonnel EL, Stephensen CB, Bennett BJ, Newman JW, Keim NL, Huang L. SNPs in apolipoproteins contribute to sex-dependent differences in blood lipids before and after a high-fat dietary challenge in healthy U.S. adults. BMC Nutr 2022; 8:95. [PMID: 36050800 PMCID: PMC9438272 DOI: 10.1186/s40795-022-00592-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 08/21/2022] [Indexed: 11/23/2022] Open
Abstract
Background The effect of genetic polymorphisms on fasting blood lipid levels have been widely studied but the effects of these within the context of a high-fat meal challenge remain less characterized. The current study aimed to investigate the association of SNPs in lipoprotein-related genes with blood lipid profiles in healthy adults in the U.S. Methods Subjects (n = 393) between 18–66 years of age with BMIs ranging from 18.5–45 kg/m2 were enrolled the cross-sectional Nutritional Phenotyping Study. Among them, 349 subjects (men: 48%; women: 52%) gave consent for genotyping. SNPs in APOA5, APOB, APOC3, APOE, and LDLR were assessed. The association between lipid markers and genotypes was tested separately for each SNP with analysis of variance (ANOVA), adjusted for sex, age, and BMI. We also examined two-factor interactions between SNPs and sex, age, or BMI. Results Women carrying the C allele of rs3135506 in APOA5 or men carrying the C allele of rs429358 in APOE had reduced HDL-cholesterol levels during fasting and postprandially. The C allele in APOE was also correlated to increased LDL-C levels. The TT genotype of rs2854116 in APOC3 was associated with elevated total cholesterol. Additive effect of the risk alleles of APOA5 and APOE or APOC3 and APOE was detected. Nevertheless, the tested SNPs had little impact on the postprandial triglyceride responses to the high-fat challenge meal. We found no significant effects of SNPs in APOB (rs1042034) or LDLR (rs2228671) on triglycerides, cholesterol, or free fatty acid levels. Conclusions In healthy adults, fasting and postprandial cholesterol levels are strongly correlated with the tested APOA5, APOE, and APOC3 genotypes. Sex contributes to the genetic impact of the tested SNPs on lipid profiles. Trial registration ClinicalTrials.gov, NCT02367287. Registered February 20, 2015, https://clinicaltrials.gov/ct2/show/NCT02367287. Supplementary Information The online version contains supplementary material available at 10.1186/s40795-022-00592-x.
Collapse
Affiliation(s)
- Yining E Wang
- Integrative Genetics and Genomics, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Catherine P Kirschke
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA
| | - Leslie R Woodhouse
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA
| | - Ellen L Bonnel
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA
| | - Charles B Stephensen
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA.,Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Brian J Bennett
- Integrative Genetics and Genomics, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA.,USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA.,Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA
| | - John W Newman
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA.,Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Nancy L Keim
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA.,Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Liping Huang
- Integrative Genetics and Genomics, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA. .,USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA. .,Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA.
| |
Collapse
|
8
|
Strahlhofer-Augsten M, Schliefsteiner C, Cvitic S, George M, Lang-Olip I, Hirschmugl B, Marsche G, Lang U, Novakovic B, Saffery R, Desoye G, Wadsack C. The Distinct Role of the HDL Receptor SR-BI in Cholesterol Homeostasis of Human Placental Arterial and Venous Endothelial Cells. Int J Mol Sci 2022; 23:ijms23105364. [PMID: 35628180 PMCID: PMC9141204 DOI: 10.3390/ijms23105364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022] Open
Abstract
As opposed to adults, high-density lipoprotein (HDL) is the main cholesterol carrying lipoprotein in fetal circulation. The major HDL receptor, scavenger receptor class B type I (SR-BI), contributes to local cholesterol homeostasis. Arterial endothelial cells (ECA) from human placenta are enriched with cholesterol compared to venous endothelial cells (ECV). Moreover, umbilical venous and arterial plasma cholesterol levels differ markedly. We tested the hypothesis that the uptake of HDL-cholesteryl esters differs between ECA and ECV because of the differential expression of SR-BI. We aimed to identify the key regulators underlying these differences and the functional consequences. Immunohistochemistry was used for visualization of SR-BI in situ. ECA and ECV were isolated from the chorionic plate of human placenta and used for RT-qPCR, Western Blot, and HDL uptake assays with 3H- and 125I-labeled HDL. DNA was extracted for the methylation profiling of the SR-BI promoter. SR-BI regulation was studied by exposing ECA and ECV to differential oxygen concentrations or shear stress. Our results show elevated SR-BI expression and protein abundance in ECA compared to ECV in situ and in vitro. Immunohistochemistry demonstrated that SR-BI is mainly expressed on the apical side of placental endothelial cells in situ, allowing interaction with mature HDL circulating in the fetal blood. This was functionally linked to a higher increase of selective cholesterol ester uptake from fetal HDL in ECA than in ECV, and resulted in increased cholesterol availability in ECA. SR-BI expression on ECV tended to decrease with shear stress, which, together with heterogeneous immunostaining, suggests that SR-BI expression is locally regulated in the placental vasculature. In addition, hypomethylation of several CpG sites within the SR-BI promoter region might contribute to differential expression of SR-BI between chorionic arteries and veins. Therefore, SR-BI contributes to a local cholesterol homeostasis in ECA and ECV of the human feto-placental vasculature.
Collapse
Affiliation(s)
- Manuela Strahlhofer-Augsten
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- BioBank Graz, Medical University of Graz, 8036 Graz, Austria
| | - Carolin Schliefsteiner
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Silvija Cvitic
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Meekha George
- Otto Loewi Research Center, Division of Pathophysiology and Immunology, Medical University of Graz, 8010 Graz, Austria;
| | - Ingrid Lang-Olip
- Gottfried Schatz Research Center, Divison of Cell Biology, Histology and Embryology, Medical University of Graz, 8036 Graz, Austria;
| | - Birgit Hirschmugl
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria;
| | - Uwe Lang
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Boris Novakovic
- Molecular Immunity, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (B.N.); (R.S.)
| | - Richard Saffery
- Molecular Immunity, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (B.N.); (R.S.)
| | - Gernot Desoye
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Christian Wadsack
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- Correspondence:
| |
Collapse
|
9
|
Luquero A, Vilahur G, Casani L, Badimon L, Borrell-Pages M. Differential cholesterol uptake in liver cells: A role for PCSK9. FASEB J 2022; 36:e22291. [PMID: 35344222 DOI: 10.1096/fj.202101660rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/11/2022] [Accepted: 03/20/2022] [Indexed: 12/21/2022]
Abstract
The clearance of low-density lipoprotein (LDL) particles from the circulation is regulated by the LDL receptor (LDLR) and proprotein convertase subtilisin/kexin 9 (PCSK9) interaction. Its disruption reduces blood cholesterol levels and delays atherosclerosis progression. Whether other members of the LDLR superfamily are in vivo targets of PCSK9 has been poorly explored. The aim of this work was to study the interaction between PCSK9 and members of the LDLR superfamily in the regulation of liver cholesterol homeostasis in an in vivo low-density lipoprotein receptor related protein 5 (LRP5) deficient mice model challenged with high-fat diet. Our results show that Wt and Lrp5-/- mice fed a hypercholesterolemic diet (HC) have increased cholesterol ester accumulation and decreased liver LDLR and LRP5 gene and protein expression. Very low-density lipoprotein receptor (VLDLR), LRP6, LRP2, and LRP1 expression levels were analyzed in liver samples and show that they do not participate in Lrp5-/- liver cholesterol uptake. Immunoprecipitation experiments show that LRP5 forms a complex with PCSK9 in liver-specific fat-storing stellate cells but not in structural HepG2 cells. Hepatic stellate cells silenced for LRP5 and/or PCSK9 expression and challenged with lipids show reduced cholesterol ester accumulation, indicating that both proteins are involved in lipid processing in the liver. Our results indicate that cholesterol esters accumulate in livers of Wt mice in a LDLR-family-members dependent manner as VLDLR, LRP2, and LRP6 show increased expression in HC mice. However, this increase is lost in livers of Lrp5-/- mice, where scavenger receptors are involved in cholesterol uptake. PCSK9 expression is strongly downregulated in mice livers after HC feeding. However PCSK9 and LRP5 bind in the cytoplasm of fat storing liver cells, indicating that this PCSK9-LRP5 interaction is cell-type specific and that both proteins contribute to lipid uptake.
Collapse
Affiliation(s)
- Aureli Luquero
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain.,CIBER-CV, Instituto de Salud Carlos III, Spain
| | - Laura Casani
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain.,CIBER-CV, Instituto de Salud Carlos III, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain.,CIBER-CV, Instituto de Salud Carlos III, Spain
| |
Collapse
|
10
|
Control of Cholesterol Metabolism Using a Systems Approach. BIOLOGY 2022; 11:biology11030430. [PMID: 35336806 PMCID: PMC8945167 DOI: 10.3390/biology11030430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 11/25/2022]
Abstract
Simple Summary Cholesterol is the main sterol in mammals that is essential for healthy cell functionining. It plays a key role in metabolic regulation and signaling, it is a precursor molecule of bile acids, oxysterols, and all steroid hormones. It also contributes to the structural makeup of the membranes. Its homeostasis is tightly controlled since it can harm the body if it is allowed to reach abnormal blood concentrations. One of the diseases associated with elevated cholesterol levels being the major cause of morbidities and mortalities worldwide, is atherosclerosis. In this study, we have developed a model of the cholesterol metabolism taking into account local inflammation and oxidative stress. The aim was to investigate the impact of the interplay of those processes and cholesterol metabolism disturbances on the atherosclerosis development and progression. We have also analyzed the effect of combining different classes of drugs targeting selected components of cholesterol metabolism. Abstract Cholesterol is an essential component of mammalian cells and is involved in many fundamental physiological processes; hence, its homeostasis in the body is tightly controlled, and any disturbance has serious consequences. Disruption of the cellular metabolism of cholesterol, accompanied by inflammation and oxidative stress, promotes the formation of atherosclerotic plaques and, consequently, is one of the leading causes of death in the Western world. Therefore, new drugs to regulate disturbed cholesterol metabolism are used and developed, which help to control cholesterol homeostasis but still do not entirely cure atherosclerosis. In this study, a Petri net-based model of human cholesterol metabolism affected by a local inflammation and oxidative stress, has been created and analyzed. The use of knockout of selected pathways allowed us to observe and study the effect of various combinations of commonly used drugs on atherosclerosis. The analysis results led to the conclusion that combination therapy, targeting multiple pathways, may be a fundamental concept in the development of more effective strategies for the treatment and prevention of atherosclerosis.
Collapse
|
11
|
Jiang XX, Bian W, Zhu YR, Wang Z, Ye P, Gu Y, Zhang H, Zuo G, Li X, Zhu L, Liu Z, Sun C, Chen SL, Zhang DM. Targeting the KCa3.1 channel suppresses diabetes-associated atherosclerosis via the STAT3/CD36 axis. Diabetes Res Clin Pract 2022; 185:109776. [PMID: 35149165 DOI: 10.1016/j.diabres.2022.109776] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 01/21/2022] [Accepted: 02/07/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND In diet-induced arterial atherosclerosis, increased KCa3.1 channel was associated with atherosclerotic plaque progression and instability. Macrophages are involved in the formation of atherosclerotic plaques, and the release of inflammatory cytokines and oxygen free radicals promotes plaque progression. However, whether the macrophage KCa3.1 channel facilitates diabetes-accelerated atherosclerosis is still unclear. This study investigated atherosclerotic plaque in ApoE-/- mice regulated by the KCa3.1 channel. METHODS AND RESULTS In vivo, blocking KCa3.1channel inhibit the development of the atherosclerotic lesion in diabetic ApoE-/- mice fed with a high-fat diet. In vitro, upregulation of KCa3.1 channel level occurred in RAW264.7 cells treated with HG plus ox-LDL in a time-dependent manner. Blocking KCa3.1 significantly reduced the uptake of ox-LDL in mice peritoneal macrophages. Further studies indicated the KCa3.1 siRNA and TRAM-34 (KCa3.1 inhibitor) attenuated the scavenger receptor CD36 expression via inhibiting STAT3 phosphorylation. CONCLUSION Blockade of macrophage KCa3.1 channel inhibit cellular oxidized low-density lipoprotein accumulation and decrease proinflammation factors expression via STAT3/CD36 axis. This study provided a novel therapeutic target to reduce the risk of atherosclerosis development in diabetic patients.
Collapse
Affiliation(s)
- Xiao-Xin Jiang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Weikang Bian
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Yan-Rong Zhu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Zhicheng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Peng Ye
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Guangfeng Zuo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Xiaobo Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Linlin Zhu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Zhizhong Liu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Chongxiu Sun
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China.
| | - Shao-Liang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China.
| | - Dai-Min Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China; Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, No. 109 Longmian Road, Nanjing 211166, PR China.
| |
Collapse
|
12
|
HDL and Lipid Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:49-61. [DOI: 10.1007/978-981-19-1592-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Li W, Du R, Xia C, Zhang H, Xie Y, Gao X, Ouyang Y, Yin Z, Hu G. Novel pituitary actions of GnRH in teleost: The link between reproduction and feeding regulation. Front Endocrinol (Lausanne) 2022; 13:982297. [PMID: 36303873 PMCID: PMC9595134 DOI: 10.3389/fendo.2022.982297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH), as a vital hypothalamic neuropeptide, was a key regulator for pituitary luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in the vertebrate. However, little is known about the other pituitary actions of GnRH in teleost. In the present study, two GnRH variants (namely, GnRH2 and GnRH3) and four GnRH receptors (namely, GnRHR1, GnRHR2, GnRHR3, and GnRHR4) had been isolated from grass carp. Tissue distribution displayed that GnRHR4 was more highly detected in the pituitary than the other three GnRHRs. Interestingly, ligand-receptor selectivity showed that GnRHR4 displayed a similar and high binding affinity for grass carp GnRH2 and GnRH3. Using primary culture grass carp pituitary cells as model, we found that both GnRH2 and GnRH3 could not only significantly induce pituitary reproductive hormone gene (GtHα, LHβ, FSHβ, INHBa, secretogranin-2) mRNA expression mediated by AC/PKA, PLC/IP3/PKC, and Ca2+/CaM/CaMK-II pathways but also reduce dopamine receptor 2 (DRD2) mRNA expression via the Ca2+/CaM/CaMK-II pathway. Interestingly, GnRH2 and GnRH3 could also stimulate anorexigenic peptide (POMCb, CART2, UTS1, NMBa, and NMBb) mRNA expression via AC/PKA, PLC/IP3/PKC, and Ca2+/CaM/CaMK-II pathways in grass carp pituitary cells. In addition, food intake could significantly induce brain GnRH2 mRNA expression. These results indicated that GnRH should be the coupling factor to integrate the feeding metabolism and reproduction in teleost.
Collapse
Affiliation(s)
- Wei Li
- Hubei Province Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Ruixin Du
- Hubei Province Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Chuanhui Xia
- Hubei Province Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Huiying Zhang
- Hubei Province Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Yunyi Xie
- Hubei Province Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Xiaowen Gao
- Hubei Province Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Yu Ouyang
- Hubei Province Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Zhan Yin, ; Guangfu Hu,
| | - Guangfu Hu
- Hubei Province Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- *Correspondence: Zhan Yin, ; Guangfu Hu,
| |
Collapse
|
14
|
Yu H. HDL and Scavenger Receptor Class B Type I (SRBI). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:79-93. [DOI: 10.1007/978-981-19-1592-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Gruber EJ, Aygun AY, Leifer CA. Macrophage uptake of oxidized and acetylated low-density lipoproteins and generation of reactive oxygen species are regulated by linear stiffness of the growth surface. PLoS One 2021; 16:e0260756. [PMID: 34914760 PMCID: PMC8675690 DOI: 10.1371/journal.pone.0260756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/16/2021] [Indexed: 01/18/2023] Open
Abstract
Macrophages are key players in the development of atherosclerosis: they scavenge lipid, transform into foam cells, and produce proinflammatory mediators. At the same time, the arterial wall undergoes profound changes in its mechanical properties. We recently showed that macrophage morphology and proinflammatory potential are regulated by the linear stiffness of the growth surface. Here we asked whether linear stiffness also regulates lipid uptake by macrophages. We cultured murine bone marrow-derived macrophages (BMMs) on polyacrylamide gels modeling stiffness of healthy (1kPa) and diseased (10-150kPa) blood vessels. In unprimed BMMs, increased linear stiffness increased uptake of oxidized (oxLDL) and acetylated (acLDL) low density lipoproteins and generation of reactive oxygen species, but did not alter phagocytosis of bacteria or silica particles. Macrophages adapted to stiff growth surfaces had increased mRNA and protein expression of two key lipoprotein receptors: CD36 and scavenger receptor b1. Regulation of the lipoprotein receptor, lectin-like receptor for ox-LDL, was more complex: mRNA expression decreased but surface protein expression increased with increased stiffness. Focal adhesion kinase was required for maximal uptake of oxLDL, but not of acLDL. Uptake of oxLDL and acLDL was independent of rho-associated coiled coil kinase. Through pharmacologic inhibition and genetic deletion, we found that transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel, plays an inhibitory role in the uptake of acLDL, but not oxLDL. Together, these results implicate mechanical signaling in the uptake of acLDL and oxLDL, opening up the possibility of new pharmacologic targets to modulate lipid uptake by macrophages in vivo.
Collapse
Affiliation(s)
- Erika J. Gruber
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Ali Y. Aygun
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Cynthia A. Leifer
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
16
|
Circadian regulation of apolipoprotein gene expression affects testosterone production in mouse testis. Theriogenology 2021; 174:9-19. [PMID: 34416563 DOI: 10.1016/j.theriogenology.2021.06.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 06/07/2021] [Accepted: 06/17/2021] [Indexed: 01/02/2023]
Abstract
The circadian clock system plays an important role in regulating testosterone synthesis in mammals. Male Bmal1-/- mice are infertile with low serum testosterone levels and decreased expression of testicular steroidogenic genes, suggesting that circadian clock genes regulate testosterone biosynthesis by activating steroidogenic gene transcription. However, whether the circadian clock regulates testosterone production via other genes remains unknown. Using Bmal1-/- mice and their wild-type (WT) siblings, we aimed to identify additional genes by which the circadian clock regulates testosterone synthesis. WT and Bmal1-/- mouse testes sections had similar normal morphologies, although there was a decrease in testicular spermatozoa in the Bmal1-/- mice. Low serum testosterone levels were detected in the Bmal1-/- mice. RNA sequencing identified 37 and 48 genes that were differentially expressed between WT and Bmal1-/- mouse testes at circadian time (CT2 and CT14), respectively. The cholesterol metabolism pathway was significantly enriched in the KEGG pathway analysis, and there was lower expression of three apolipoprotein genes (Apoa1, Apoa2, and Apoc3) at CT2 in the testes of Bmal1-/- mice than in those of WT mice. These decreases in Apoa1, Apoa2, and Apoc3 expression were verified by quantitative polymerase chain reaction analysis, which also revealed downregulation of the expression of the circadian clock (Per2, Dbp, and Nr1d1) and steroidogenic (StAR, Cyp11a1, and Hsd17b3) genes. The expression of circadian clock genes was relatively stable in WT mice over a 20-h period, whereas there was clear circadian rhythmic expression of Apoa1, Apoa2, Apoc3, StAR, Cyp11a1, Hsd3b2, and Hsd17b3. Bmal1-/- mice showed severely reduced expression of testicular circadian clock genes at three time points (CT4, CT12, and CT20), and a reduction in mRNA expression levels of Apo (Apoa1, Apoa2, and Apoc3) and steroidogenic (StAR, Cyp11a1, Hsd3b2, and Hsd17b3) genes. Oil Red O staining showed decreased lipid aggregation in the Leydig cells of Bmal1-/- mouse testes. Considering the vital role of Apo genes in high-density lipoprotein formation and cholesterol transport, the present data suggest that the circadian clock system regulates testosterone production by orchestrating the rhythmic expression of Apo genes. These data extend our understanding of the role of the circadian clock in regulating testosterone production in mammals.
Collapse
|
17
|
Kluck GEG, Yoo JA, Sakarya EH, Trigatti BL. Good Cholesterol Gone Bad? HDL and COVID-19. Int J Mol Sci 2021; 22:10182. [PMID: 34638523 PMCID: PMC8507803 DOI: 10.3390/ijms221910182] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
The transmissible respiratory disease COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has affected millions of people worldwide since its first reported outbreak in December of 2019 in Wuhan, China. Since then, multiple studies have shown an inverse correlation between the levels of high-density lipoprotein (HDL) particles and the severity of COVID-19, with low HDL levels being associated with an increased risk of severe outcomes. Some studies revealed that HDL binds to SARS-CoV-2 particles via the virus's spike protein and, under certain conditions, such as low HDL particle concentrations, it facilitates SARS-CoV-2 binding to angiotensin-converting enzyme 2 (ACE2) and infection of host cells. Other studies, however, reported that HDL suppressed SARS-CoV-2 infection. In both cases, the ability of HDL to enhance or suppress virus infection appears to be dependent on the expression of the HDL receptor, namely, the Scavenger Receptor Class B type 1 (SR-B1), in the target cells. SR-B1 and HDL represent crucial mediators of cholesterol metabolism. Herein, we review the complex role of HDL and SR-B1 in SARS-CoV-2-induced disease. We also review recent advances in our understanding of HDL structure, properties, and function during SARS-CoV-2 infection and the resulting COVID-19 disease.
Collapse
Affiliation(s)
| | | | | | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute and Department of Biochemistry and Biomedical Sciences, McMaster University and Hamilton Health Sciences, Hamilton, ON L8L 2X2, Canada; (G.E.G.K.); (J.-A.Y.); (E.H.S.)
| |
Collapse
|
18
|
Naito T, Saheki Y. GRAMD1-mediated accessible cholesterol sensing and transport. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158957. [PMID: 33932585 DOI: 10.1016/j.bbalip.2021.158957] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 01/19/2023]
Abstract
Cholesterol, an essential lipid for cell signaling and structural integrity of cellular membranes, is highly enriched in the plasma membrane (PM). However, the regulatory mechanisms that control its biosynthesis and uptake both reside in the endoplasmic reticulum (ER). Thus, the ER needs to constantly monitor the levels of PM cholesterol. This is in part mediated by regulated transport of a biochemically defined pool of cholesterol, termed "accessible" cholesterol, from the PM to the ER via evolutionarily conserved ER-anchored lipid transfer proteins, the GRAMD1s/Asters (GRAMD1a/1b/1c) (Lam/Ltc proteins in yeast). GRAMD1s possess cytosolically exposed GRAM domain and StART-like domain followed by a transmembrane ER anchor. They form homo- and hetero-meric complexes and move to the contacts formed between the ER and the PM by sensing a transient expansion of the accessible pool of cholesterol in the PM via the GRAM domain and facilitate its extraction and transport to the ER via the StART-like domain. The GRAMD1b GRAM domain possesses distinct, but synergistic sites, for recognizing accessible cholesterol and anionic lipids, including phosphatidylserine, within the PM. This property of the GRAM domain contributes to regulated tethering of the PM to ER membrane where GRAMD1s are anchored and fine-tunes StART-like domain-dependent accessible cholesterol transport. Thus, cells use GRAMD1s to sense the levels of cholesterol in the PM and regulate transport of accessible PM cholesterol to the ER in order to maintain cholesterol homeostasis.
Collapse
Affiliation(s)
- Tomoki Naito
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore; Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan.
| |
Collapse
|
19
|
Cabodevilla AG, Tang S, Lee S, Mullick AE, Aleman JO, Hussain MM, Sessa WC, Abumrad NA, Goldberg IJ. Eruptive xanthoma model reveals endothelial cells internalize and metabolize chylomicrons, leading to extravascular triglyceride accumulation. J Clin Invest 2021; 131:e145800. [PMID: 34128469 PMCID: PMC8203467 DOI: 10.1172/jci145800] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
Although tissue uptake of fatty acids from chylomicrons is primarily via lipoprotein lipase (LpL) hydrolysis of triglycerides (TGs), studies of patients with genetic LpL deficiency suggest additional pathways deliver dietary lipids to tissues. Despite an intact endothelial cell (EC) barrier, hyperchylomicronemic patients accumulate chylomicron-derived lipids within skin macrophages, leading to the clinical finding eruptive xanthomas. We explored whether an LpL-independent pathway exists for transfer of circulating lipids across the EC barrier. We found that LpL-deficient mice had a marked increase in aortic EC lipid droplets before and after a fat gavage. Cultured ECs internalized chylomicrons, which were hydrolyzed within lysosomes. The products of this hydrolysis fueled lipid droplet biogenesis in ECs and triggered lipid accumulation in cocultured macrophages. EC chylomicron uptake was inhibited by competition with HDL and knockdown of the scavenger receptor-BI (SR-BI). In vivo, SR-BI knockdown reduced TG accumulation in aortic ECs and skin macrophages of LpL-deficient mice. Thus, ECs internalize chylomicrons, metabolize them in lysosomes, and either store or release their lipids. This latter process may allow accumulation of TGs within skin macrophages and illustrates a pathway that might be responsible for creation of eruptive xanthomas.
Collapse
Affiliation(s)
- Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Songtao Tang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Sungwoon Lee
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jose O Aleman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - M Mahmood Hussain
- Diabetes and Obesity Center, NYU-Long Island School of Medicine, Mineola, New York, USA
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nada A Abumrad
- Nutritional Sciences, Department of Medicine and Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
20
|
Abumrad NA, Cabodevilla AG, Samovski D, Pietka T, Basu D, Goldberg IJ. Endothelial Cell Receptors in Tissue Lipid Uptake and Metabolism. Circ Res 2021; 128:433-450. [PMID: 33539224 DOI: 10.1161/circresaha.120.318003] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipid uptake and metabolism are central to the function of organs such as heart, skeletal muscle, and adipose tissue. Although most heart energy derives from fatty acids (FAs), excess lipid accumulation can cause cardiomyopathy. Similarly, high delivery of cholesterol can initiate coronary artery atherosclerosis. Hearts and arteries-unlike liver and adrenals-have nonfenestrated capillaries and lipid accumulation in both health and disease requires lipid movement from the circulation across the endothelial barrier. This review summarizes recent in vitro and in vivo findings on the importance of endothelial cell receptors and uptake pathways in regulating FAs and cholesterol uptake in normal physiology and cardiovascular disease. We highlight clinical and experimental data on the roles of ECs in lipid supply to tissues, heart, and arterial wall in particular, and how this affects organ metabolism and function. Models of FA uptake into ECs suggest that receptor-mediated uptake predominates at low FA concentrations, such as during fasting, whereas FA uptake during lipolysis of chylomicrons may involve paracellular movement. Similarly, in the setting of an intact arterial endothelial layer, recent and historic data support a role for receptor-mediated processes in the movement of lipoproteins into the subarterial space. We conclude with thoughts on the need to better understand endothelial lipid transfer for fuller comprehension of the pathophysiology of hyperlipidemia, and lipotoxic diseases such as some forms of cardiomyopathy and atherosclerosis.
Collapse
Affiliation(s)
- Nada A Abumrad
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Dmitri Samovski
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Terri Pietka
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| |
Collapse
|
21
|
Yu L, Dai Y, Mineo C. Novel Functions of Endothelial Scavenger Receptor Class B Type I. Curr Atheroscler Rep 2021; 23:6. [PMID: 33420646 DOI: 10.1007/s11883-020-00903-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Scavenger receptor class B type I (SR-BI) serves a key role in the reverse cholesterol transport in the liver as the high-affinity receptor for HDL. SR-BI is abundantly expressed in endothelium, and earlier works indicate that the receptor mediates anti-atherogenic actions of HDL. However, more recent studies uncovered novel functions of endothelial SR-BI as a lipoprotein transporter, which regulates transcellular transport process of both LDL and HDL. This brief review focuses on the unique functions of endothelial SR-BI and how they influence atherogenesis. RECENT FINDINGS Earlier studies indicate that SR-BI facilitates anti-atherogenic actions of HDL through modulation of intracellular signaling to stimulate endothelial nitric oxide synthase. In vivo studies in global SR-BI knockout mice also showed a strong atheroprotective role of the receptor; however, a contribution of endothelial SR-BI to atherosclerosis process in vivo has not been fully appreciated. Recent studies using cultured endothelial cells and in mice with endothelial-specific deletion of the receptor revealed previously unappreciated pro-atherogenic actions of SR-BI, which relates to its ability to deliver LDL into arteries. On the other hand, SR-BI has also been implicated in transport of HDL to the sub-intimal space as a part of reverse cholesterol transport. SR-BI mediates internalization and transcellular transport of both HDL and LDL, and the cellular and molecular mechanism of the process has just begun to emerge. Harnessing these dual transport functions of the endothelial SR-BI may provide a novel, effective intervention to atherosclerosis.
Collapse
Affiliation(s)
- Liming Yu
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yao Dai
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
22
|
Peli1 impairs microglial Aβ phagocytosis through promoting C/EBPβ degradation. PLoS Biol 2020; 18:e3000837. [PMID: 33017390 PMCID: PMC7561136 DOI: 10.1371/journal.pbio.3000837] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 10/15/2020] [Accepted: 08/26/2020] [Indexed: 12/29/2022] Open
Abstract
Amyloid-β (Aβ) accumulation in the brain is a hallmark of Alzheimer’s disease (AD) pathology. However, the molecular mechanism controlling microglial Aβ phagocytosis is poorly understood. Here we found that the E3 ubiquitin ligase Pellino 1 (Peli1) is induced in the microglia of AD-like five familial AD (5×FAD) mice, whose phagocytic efficiency for Aβ was then impaired, and therefore Peli1 depletion suppressed the Aβ deposition in the brains of 5×FAD mice. Mechanistic characterizations indicated that Peli1 directly targeted CCAAT/enhancer-binding protein (C/EBP)β, a major transcription factor responsible for the transcription of scavenger receptor CD36. Peli1 functioned as a direct E3 ubiquitin ligase of C/EBPβ and mediated its ubiquitination-induced degradation. Consequently, loss of Peli1 increased the protein levels of C/EBPβ and the expression of CD36 and thus, promoted the phagocytic ability in microglial cells. Together, our findings established Peli1 as a critical regulator of microglial phagocytosis and highlighted the therapeutic potential by targeting Peli1 for the treatment of microglia-mediated neurological diseases. This study identifies Peli1, an E3 ubiqitin ligase enriched in microglia, as a restraining factor that curtails microglial phagocytosis of the amyloid Aβ. Correspondingly, deletion of Peli1 enhances Aβ phagocytosis and clearance in Alzheimer’s disease, implicating Peli1 as a therapeutic target with significant potential for the treatment of microglia-mediated neurological disease.
Collapse
|
23
|
Liu J, Zhao M, Zhu Y, Zheng L, Yin Y. Plasma Metabolomic and Lipidomic Profiling of a Genetically Modified Mouse Model of Scavenger Receptor Class B Type I. Proteomics 2020; 20:e2000050. [PMID: 33090674 DOI: 10.1002/pmic.202000050] [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: 03/07/2020] [Revised: 08/31/2020] [Indexed: 11/06/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall and is becoming the principal cause of death globally. The reverse cholesterol transport (RCT) mediated by scavenger receptor class B type I (SR-BI) is a major protection mechanism against atherosclerosis. To investigate the metabolome changes and to find potential biomarkers involved in RCT, nontargeted metabolomics and nontargeted lipidomics are applied to SR-BI knockout mice that are fed a high fat and high cholesterol diet. SR-BI knockout mice and controls are told apart using multidimensional statistical analysis, and potential biomarkers are found and identified. The pathophysiological meaning of the biomarkers and the perturbed metabolic pathways are also addressed, which could provide new evidence for atherosclerosis studies.
Collapse
Affiliation(s)
- Jia Liu
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Yizhang Zhu
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, 100191, China
| |
Collapse
|
24
|
Ruiz-Roso MB, Gil-Zamorano J, López de Las Hazas MC, Tomé-Carneiro J, Crespo MC, Latasa MJ, Briand O, Sánchez-López D, Ortiz AI, Visioli F, Martínez JA, Dávalos A. Intestinal Lipid Metabolism Genes Regulated by miRNAs. Front Genet 2020; 11:707. [PMID: 32742270 PMCID: PMC7366872 DOI: 10.3389/fgene.2020.00707] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) crucial roles in translation repression and post-transcriptional adjustments contribute to regulate intestinal lipid metabolism. Even though their actions in different metabolic tissues have been elucidated, their intestinal activity is yet unclear. We aimed to investigate intestinal miRNA-regulated lipid metabolism-related genes, by creating an intestinal-specific Dicer1 knockout (Int-Dicer1 KO) mouse model, with a depletion of microRNAs in enterocytes. The levels of 83 cholesterol and lipoprotein metabolism-related genes were assessed in the intestinal mucosa of Int-Dicer1 KO and Wild Type C57BL/6 (WT) littermates mice at baseline and 2 h after an oral lipid challenge. Among the 18 genes selected for further validation, Hmgcs2, Acat1 and Olr1 were found to be strong candidates to be modulated by miRNAs in enterocytes and intestinal organoids. Moreover, we report that intestinal miRNAs contribute to the regulation of intestinal epithelial differentiation. Twenty-nine common miRNAs found in the intestines were analyzed for their potential to target any of the three candidate genes found and validated by miRNA-transfection assays in Caco-2 cells. MiR-31-5p, miR-99b-5p, miR-200a-5p, miR-200b-5p and miR-425-5p are major regulators of these lipid metabolism-related genes. Our data provide new evidence on the potential of intestinal miRNAs as therapeutic targets in lipid metabolism-associated pathologies.
Collapse
Affiliation(s)
- María Belén Ruiz-Roso
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Judit Gil-Zamorano
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - María Carmen López de Las Hazas
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Joao Tomé-Carneiro
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - María Carmen Crespo
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - María Jesús Latasa
- Research Program, Innovation, Communication and Education Program, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Olivier Briand
- University of Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Daniel Sánchez-López
- University of Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Ana I Ortiz
- Servicio de Cirugía Experimental, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Francesco Visioli
- Laboratory of Functional Foods, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain.,Department of Molecular Medicine, University of Padua, Padua, Italy
| | - J Alfredo Martínez
- Department of Nutrition and Physiology, Center for Nutrition Research, University of Navarra, IDISNA Navarra, Pamplona, Spain.,Centre of Biomedical Research in Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain.,Cardiometabolic Nutrition Group, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| | - Alberto Dávalos
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute for Advanced Studies (IMDEA)-Food, CEI UAM + CSIC, Madrid, Spain
| |
Collapse
|
25
|
Shemiakova T, Ivanova E, Grechko AV, Gerasimova EV, Sobenin IA, Orekhov AN. Mitochondrial Dysfunction and DNA Damage in the Context of Pathogenesis of Atherosclerosis. Biomedicines 2020; 8:E166. [PMID: 32570831 PMCID: PMC7344998 DOI: 10.3390/biomedicines8060166] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a multifactorial disease of the cardiovascular system associated with aging, inflammation, and oxidative stress. An important role in the development of atherosclerosis play elevated plasma lipoproteins. A number of external factors (smoking, diabetes, infections) can also contribute to the development of the disease. For a long time, atherosclerosis remains asymptomatic, therefore, the search for early markers of the disease is critical for the timely management and better outcomes for patients. Mitochondrial dysfunction and mitochondrial DNA (mtDNA) damage appear to connect different aspects of atherosclerosis pathogenesis. To date, multiple lines of research have demonstrated the strong association of mitochondrial dysfunction with the development of various human diseases. Therapies aimed at restoring the mitochondrial function are being actively developed, and are expected to broaden the therapeutic possibilities for several chronic human diseases. The development of such therapies depends on our understanding of the functional roles of different mtDNA variants associated with one or another disorder, and the molecular mechanisms linking mitochondrial dysfunction with a given pathological feature. These questions are, however, challenging and require future intensive research. This review summarizes the recent studies and describes the central processes of the development of atherosclerosis, and shows their relationship with mitochondrial dysfunction. One of the promising therapeutic approaches for future atherosclerosis treatments is the use of mitochondria-targeted antioxidants. Future studies should focus on characterizing the mechanisms of mitochondrial involvement in cardiovascular pathologies to better direct the search for novel therapies.
Collapse
Affiliation(s)
- Taisiia Shemiakova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia;
| | - Ekaterina Ivanova
- Department of Basic Research, Institute for Atherosclerosis Research, 121609 Moscow, Russia
| | - Andrey V. Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, 109240 Moscow, Russia;
| | - Elena V. Gerasimova
- Laboratory of Systemic Rheumatic Disorders, V.A. Nasonova Institute of Rheumatology, 115522 Moscow, Russia;
| | - Igor A. Sobenin
- Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 121552 Moscow, Russia;
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Laboratory of Infection Pathology and Molecular Microecology, Institute of Human Morphology, 117418 Moscow, Russia
| |
Collapse
|
26
|
Li S, Xu S, Zhao Y, Wang H, Feng J. Dietary Betaine Addition Promotes Hepatic Cholesterol Synthesis, Bile Acid Conversion, and Export in Rats. Nutrients 2020; 12:nu12051399. [PMID: 32414094 PMCID: PMC7284822 DOI: 10.3390/nu12051399] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 12/25/2022] Open
Abstract
It is widely reported how betaine addition regulates lipid metabolism but how betaine affects cholesterol metabolism is still unknown. This study aimed to investigate the role of betaine in hepatic cholesterol metabolism of Sprague-Dawley rats. Rats were randomly allocated to four groups and fed with a basal diet or a high-fat diet with or without 1% betaine. The experiment lasted 28 days. The results showed that dietary betaine supplementation reduced the feed intake of rats with final weight unchanged. Serum low-density-lipoprotein cholesterol was increased with the high-fat diet. The high-fat diet promoted cholesterol synthesis and excretion by enhancing the HMG-CoA reductase and ABCG5/G8, respectively, which lead to a balance of hepatic cholesterol. Rats in betaine groups showed a higher level of hepatic total cholesterol. Dietary betaine addition enhanced cholesterol synthesis as well as conversion of bile acid from cholesterol by increasing the levels of HMGCR and CYP7A1. The high-fat diet decreased the level of bile salt export pump, while dietary betaine addition inhibited this decrease and promoted bile acid efflux and increased total bile acid levels in the intestine. In summary, dietary betaine addition promoted hepatic cholesterol metabolism, including cholesterol synthesis, conversion of bile acids, and bile acid export.
Collapse
Affiliation(s)
- Sisi Li
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (S.L.); (S.X.); (Y.Z.)
| | - Shuyi Xu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (S.L.); (S.X.); (Y.Z.)
| | - Yang Zhao
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (S.L.); (S.X.); (Y.Z.)
| | - Haichao Wang
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 430068, China;
| | - Jie Feng
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (S.L.); (S.X.); (Y.Z.)
- Correspondence: ; Tel.: +86-571-88982121
| |
Collapse
|
27
|
Nishida M, Takeno S, Takemoto K, Takahara D, Hamamoto T, Ishino T, Kawasumi T. Increased Tissue Expression of Lectin-Like Oxidized LDL Receptor-1 (LOX-1) Is Associated with Disease Severity in Chronic Rhinosinusitis with Nasal Polyps. Diagnostics (Basel) 2020; 10:diagnostics10040246. [PMID: 32340234 PMCID: PMC7236656 DOI: 10.3390/diagnostics10040246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background: The oxidative stress, induced by both environmental and intrinsic stimuli, underlies the onset and persistency of chronic rhinosinusitis (CRS). Scavenger receptors (SRs) are a broad family of transmembrane receptors involved in a dysfunctional host–environment interaction through a reaction with reactive oxygen species (ROS) production. Objective: We hypothesized possible roles of two major SRs in CRS pathology that can translate to clinical phenotypes or histological subtypes: lectin-like oxidized low-density lipoproteins (LDL) receptor-1 (LOX-1) and scavenger receptor class B type 1 (SR-B1). Patients and Methods: We collected ethmoid sinus mucosa specimens and blood samples from patients with CRS with nasal polyps (CRSwNP; n = 31) or CRS without NP (CRSsNP; n = 13) and 19 control subjects. We performed an RT-PCR analysis, ELISA assay, and immunostaining to determine the expressions and distributions of LOX-1 and SR-B1. Results: The CRSwNP group showed a significant increase in LOX-1 mRNA expression compared to the control group. There was no significant difference in SR-B1 mRNA levels among the three groups. The LOX-1 mRNA levels were positively correlated with the sinus computed tomography (CT) scores. Sinus tissue, but not serum samples, showed elevated concentrations of LOX-1 protein in the CRSwNP group versus the control group. The LOX-1 protein distribution was localized in inflammatory cells and vascular endothelial cells. Conclusion: LOX-1 is a major receptor for oxidized low-density lipoprotein produced by oxidative stress. This is the first study to report alterations in LOX-1 expression and production triggered by persistent inflammatory processes in CRSwNP patients. Our findings reveal complex but important roles for SRs that may contribute to the onset of different CRS phenotypes.
Collapse
Affiliation(s)
- Manabu Nishida
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan
| | - Sachio Takeno
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan
| | - Kohta Takemoto
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan
| | - Daisuke Takahara
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan
| | - Takao Hamamoto
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan
| | - Takashi Ishino
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan
| | - Tomohiro Kawasumi
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan
| |
Collapse
|
28
|
Identification of two major autoantigens negatively regulating endothelial activation in Takayasu arteritis. Nat Commun 2020; 11:1253. [PMID: 32152303 PMCID: PMC7062749 DOI: 10.1038/s41467-020-15088-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/17/2020] [Indexed: 12/19/2022] Open
Abstract
The presence of antiendothelial cell antibodies (AECAs) has been documented in Takayasu arteritis (TAK), a chronic granulomatous vasculitis. Here, we identify cell-surface autoantigens using an expression cloning system. A cDNA library of endothelial cells is retrovirally transfected into a rat myeloma cell line from which AECA-positive clones are sorted with flow cytometry. Four distinct AECA-positive clones are isolated, and endothelial protein C receptor (EPCR) and scavenger receptor class B type 1 (SR-BI) are identified as endothelial autoantigens. Autoantibodies against EPCR and SR-BI are detected in 34.6% and 36.5% of cases, respectively, with minimal overlap (3.8%). Autoantibodies against EPCR are also detected in ulcerative colitis, the frequent comorbidity of TAK. In mechanistic studies, EPCR and SR-BI function as negative regulators of endothelial activation. EPCR has also an effect on human T cells and impair Th17 differentiation. Autoantibodies against EPCR and SR-BI block the functions of their targets, thereby promoting pro-inflammatory phenotype. Autoantibodies against endothelium have been recognized in Takayasu arteritis (TAK). Here the authors identify endothelial protein C receptor and scavenger receptor class B type 1 as major autoantigens in TAK, and find autoantibodies inhibit the negative regulation of endothelial activation.
Collapse
|
29
|
Atherosclerosis and the Capillary Network; Pathophysiology and Potential Therapeutic Strategies. Cells 2019; 9:cells9010050. [PMID: 31878229 PMCID: PMC7016600 DOI: 10.3390/cells9010050] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis and associated ischemic organ dysfunction represent the number one cause of mortality worldwide. While the key drivers of atherosclerosis, arterial hypertension, hypercholesterolemia and diabetes mellitus, are well known disease entities and their contribution to the formation of atherosclerotic plaques are intensively studied and well understood, less effort is put on the effect of these disease states on microvascular structure an integrity. In this review we summarize the pathological changes occurring in the vascular system in response to prolonged exposure to these major risk factors, with a particular focus on the differences between these pathological alterations of the vessel wall in larger arteries as compared to the microcirculation. Furthermore, we intend to highlight potential therapeutic strategies to improve microvascular function during atherosclerotic vessel disease.
Collapse
|
30
|
Martins Cardoso R, Creemers E, Absalah S, Hoekstra M, Gooris GS, Bouwstra JA, Van Eck M. Hyperalphalipoproteinemic scavenger receptor BI knockout mice exhibit a disrupted epidermal lipid barrier. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158592. [PMID: 31863970 DOI: 10.1016/j.bbalip.2019.158592] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/05/2019] [Accepted: 12/16/2019] [Indexed: 01/28/2023]
Abstract
Scavenger receptor class B type I (SR-BI) mediates the selective uptake of cholesteryl esters (CE) from high-density lipoproteins (HDL). An impaired SR-BI function leads to hyperalphalipoproteinemia with elevated levels of cholesterol transported in the HDL fraction. Accumulation of cholesterol in apolipoprotein B (apoB)-containing lipoproteins has been shown to alter skin lipid composition and barrier function in mice. To investigate whether these hypercholesterolemic effects on the skin also occur in hyperalphalipoproteinemia, we compared skins of wild-type and SR-BI knockout (SR-BI-/-) mice. SR-BI deficiency did not affect the epidermal cholesterol content and induced only minor changes in the ceramide subclasses. The epidermal free fatty acid (FFA) pool was, however, enriched in short and unsaturated chains. Plasma CE levels strongly correlated with epidermal FFA C18:1 content. The increase in epidermal FFA coincided with downregulation of cholesterol and FFA synthesis genes, suggesting a compensatory response to increased flux of plasma cholesterol and FFAs into the skin. Importantly, the SR-BI-/- epidermal lipid barrier showed increased permeability to ethyl-paraminobenzoic acid, indicating an impairment of the barrier function. In conclusion, increased HDL-cholesterol levels in SR-BI-/- mice can alter the epidermal lipid composition and lipid barrier function similarly as observed in hypercholesterolemia due to elevated levels of apoB-containing lipoproteins.
Collapse
Affiliation(s)
- Renata Martins Cardoso
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Zuid-Holland, the Netherlands.
| | - Eline Creemers
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Zuid-Holland, the Netherlands
| | - Samira Absalah
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Zuid-Holland, the Netherlands.
| | - Menno Hoekstra
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Zuid-Holland, the Netherlands.
| | - Gert S Gooris
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Zuid-Holland, the Netherlands.
| | - Joke A Bouwstra
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Zuid-Holland, the Netherlands.
| | - Miranda Van Eck
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Zuid-Holland, the Netherlands.
| |
Collapse
|
31
|
Cruz S, Narayanaswami V. Cellular Uptake and Clearance of Oxidatively-modified Apolipoprotein E3 by Cerebral Cortex Endothelial Cells. Int J Mol Sci 2019; 20:ijms20184582. [PMID: 31533203 PMCID: PMC6769588 DOI: 10.3390/ijms20184582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/12/2022] Open
Abstract
Apolipoprotein E3 (apoE3) plays a critical role in the metabolism of lipoproteins and lowers plasma lipid levels by serving as a ligand for the low-density lipoprotein receptor (LDLr) family of proteins and by promoting macrophage cholesterol efflux. The current study examines the effect of acrolein (an endogenously generated metabolite and an environmental pollutant) modification on the structure and function of apoE3. Acrolein modification was confirmed in Western blots by reactivity with acrolein–lysine-specific antibody and by the presence of oligomeric species due to cross-linking. LC-MS/MS analysis revealed modification of 10 out of 12 lysines in apoE3, with Nε-(3-methylpyridinium)-lysine being the predominant form of modification, and Lys75 being a ‘hot spot’ in terms of susceptibility to oxidation. Circular dichroism spectroscopy showed no major change in overall secondary structure compared to unmodified apoE3. Reconstituted high density lipoprotein (HDL) bearing acrolein modified apoE3 showed loss of binding to soluble LDLr; however, incubation with mouse endothelioma bEnd.3 cells showed that it was internalized. Incubation with excess LDL did not abolish cellular uptake of acrolein modified apoE3, suggesting alternative mechanism(s) not involving LDLr. Incubation with anti-CD36 antibody did not show a decrease in internalization while incubation with anti- lectin-like oxidized LDL receptor 1 (LOX1) showed partial internalization. However, incubation with anti-scavenger receptor class B type I (SRB1) antibody abolished internalization of acrolein modified apoE3. Taken together, our studies suggest that acrolein modification of apoE3 at lysine residues leads to increase in net negative charge, and as a consequence, results in clearance by LOX1 and SRB1 on endothelial cells. Overall, oxidative modification of apoE3 likely impairs its role in regulating plasma cholesterol homeostasis, eventually leading to lipid disorders.
Collapse
Affiliation(s)
- Siobanth Cruz
- Department of Chemistry and Biochemistry 1250 Bellflower Blvd., California State University Long Beach, Long Beach, CA 90840, USA.
| | - Vasanthy Narayanaswami
- Department of Chemistry and Biochemistry 1250 Bellflower Blvd., California State University Long Beach, Long Beach, CA 90840, USA.
| |
Collapse
|
32
|
Zheng Z, Ai J, Guo L, Ye X, Bondada S, Howatt D, Daugherty A, Li XA. SR-BI (Scavenger Receptor Class B Type 1) Is Critical in Maintaining Normal T-Cell Development and Enhancing Thymic Regeneration. Arterioscler Thromb Vasc Biol 2019; 38:2706-2717. [PMID: 30354229 DOI: 10.1161/atvbaha.118.311728] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- Continuous T-cell production from thymus is essential in replenishing naïve T-cell pool and maintaining optimal T-cell functions. However, the underlying mechanisms regulating the T-cell development in thymus remains largely unknown. Approach and Results- We identified SR-BI (scavenger receptor class B type 1), an HDL (high-density lipoprotein) receptor, as a novel modulator in T-cell development. We found that SR-BI deficiency in mice led to reduced thymus size and decreased T-cell production, which was accompanied by narrowed peripheral naïve T-cell pool. Further investigation revealed that SR-BI deficiency impaired progenitor thymic homing, causing a dramatic reduction in the percentage of earliest thymic progenitors, but did not affect other downstream T-cell developmental steps inside the thymus. As a result of the impaired progenitor thymic homing, SR-BI-deficient mice displayed delayed thymic regeneration postirradiation. Using a variety of experimental approaches, we revealed that the impaired T-cell development in SR-BI-deficient mice was not caused by hematopoietic SR-BI deficiency or SR-BI deficiency-induced hypercholesterolemia, but mainly attributed to the SR-BI deficiency in adrenal glands, as adrenal-specific SR-BI-deficient mice exhibited similar defects in T-cell development and thymic regeneration with SR-BI-deficient mice. Conclusions- This study demonstrates that SR-BI deficiency impaired T-cell development and delayed thymic regeneration by affecting progenitor thymic homing in mice, elucidating a previously unrecognized link between SR-BI and adaptive immunity.
Collapse
Affiliation(s)
- Zhong Zheng
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Junting Ai
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Ling Guo
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Xiang Ye
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Subbarao Bondada
- Department of Microbiology (S.B.), University of Kentucky College of Medicine, Lexington
| | - Deborah Howatt
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Alan Daugherty
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Department of Physiology (A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Xiang-An Li
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Department of Physiology (A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| |
Collapse
|
33
|
Gordon JA, Noble JW, Midha A, Derakhshan F, Wang G, Adomat HH, Tomlinson Guns ES, Lin YY, Ren S, Collins CC, Nelson PS, Morrissey C, Wasan KM, Cox ME. Upregulation of Scavenger Receptor B1 Is Required for Steroidogenic and Nonsteroidogenic Cholesterol Metabolism in Prostate Cancer. Cancer Res 2019; 79:3320-3331. [PMID: 31064850 PMCID: PMC6606386 DOI: 10.1158/0008-5472.can-18-2529] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/25/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Abstract
Aberrant cholesterol metabolism is increasingly appreciated to be essential for prostate cancer initiation and progression. Transcript expression of the high-density lipoprotein-cholesterol receptor scavenger receptor B1 (SR-B1) is elevated in primary prostate cancer. Hypothesizing that SR-B1 expression may help facilitate malignant transformation, we document increased SR-B1 protein and transcript expression in prostate cancer relative to normal prostate epithelium that persists in lethal castration-resistant prostate cancer (CRPC) metastasis. As intratumoral steroid synthesis from the precursor cholesterol can drive androgen receptor (AR) pathway activity in CRPC, we screened androgenic benign and cancer cell lines for sensitivity to SR-B1 antagonism. Benign cells were insensitive to SR-B1 antagonism, and cancer line sensitivity inversely correlated with expression levels of full-length and splice variant AR. In androgen-responsive CRPC cell model C4-2, SR-B1 antagonism suppressed cholesterol uptake, de novo steroidogenesis, and AR activity. SR-B1 antagonism also suppressed growth and viability and induced endoplasmic reticulum stress and autophagy. The inability of exogenous steroids to reverse these effects indicates that AR pathway activation is insufficient to overcome cytotoxic stress caused by a decrease in the availability of cholesterol. Furthermore, SR-B1 antagonism decreased cholesterol uptake, growth, and viability of the AR-null CRPC cell model PC-3, and the small-molecule SR-B1 antagonist block lipid transport-1 decreased xenograft growth rate despite poor pharmacologic properties. Overall, our findings show that SR-B1 is upregulated in primary and castration-resistant disease and is essential for cholesterol uptake needed to drive both steroidogenic and nonsteroidogenic biogenic pathways, thus implicating SR-B1 as a novel and potentially actionable target in CRPC. SIGNIFICANCE: These findings highlight SR-B1 as a potential target in primary and castration-resistant prostate cancer that is essential for cholesterol uptake needed to drive steroidogenic and nonsteroidogenic biogenic pathways.
Collapse
MESH Headings
- Androgens/metabolism
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Bone Neoplasms/metabolism
- Bone Neoplasms/secondary
- Bone Neoplasms/surgery
- Cell Proliferation
- Cholesterol/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Liver Neoplasms/surgery
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lung Neoplasms/surgery
- Male
- Mice
- Mice, Nude
- Orchiectomy
- Prognosis
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/surgery
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jacob A Gordon
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Jake W Noble
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Ankur Midha
- Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Fatemeh Derakhshan
- Department of Pathology, British Columbia Cancer Agency, Vancouver, Canada
| | - Gang Wang
- Department of Pathology, British Columbia Cancer Agency, Vancouver, Canada
| | - Hans H Adomat
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Emma S Tomlinson Guns
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Yen-Yi Lin
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Shancheng Ren
- Department of Urology, Second Military Medical University, Shanghai, China
| | - Collin C Collins
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Kishor M Wasan
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Michael E Cox
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
- Department of Urologic Sciences, University of British Columbia, Canada
| |
Collapse
|
34
|
Xepapadaki E, Zvintzou E, Kalogeropoulou C, Filou S, Kypreos KE. Τhe Antioxidant Function of HDL in Atherosclerosis. Angiology 2019; 71:112-121. [PMID: 31185723 DOI: 10.1177/0003319719854609] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Atherosclerosis is a multistep process that progresses over a long period of time and displays a broad range of severity. In its final form, it manifests as a lesion of the intimal layer of the arterial wall. There is strong evidence supporting that oxidative stress contributes to coronary heart disease morbidity and mortality and antioxidant high-density lipoprotein (HDL) could have a beneficial role in the prevention and prognosis of the disease. Indeed, certain subspecies of HDL may act as natural antioxidants preventing oxidation of lipids on low-density lipoprotein (LDL) and biological membranes. The antioxidant function may be attributed to inhibition of synthesis or neutralization of free radicals and reactive oxygen species by HDL lipids and associated enzymes or transfer of oxidation prone lipids from LDL and biological membranes to HDL for catabolism. A limited number of clinical trials suggest that the increased antioxidant potential of HDL correlates with decreased risk for atherosclerosis. Some nutritional interventions to increase HDL antioxidant activity have been proposed with limited success so far. The limitations in measuring and understanding HDL antioxidant function in vivo are also discussed.
Collapse
Affiliation(s)
- Eva Xepapadaki
- Department of Pharmacology, School of Medicine, University of Patras, Rio Achaias, TK, Greece
| | - Evangelia Zvintzou
- Department of Pharmacology, School of Medicine, University of Patras, Rio Achaias, TK, Greece
| | | | - Serafoula Filou
- Department of Pharmacology, School of Medicine, University of Patras, Rio Achaias, TK, Greece
| | - Kyriakos E Kypreos
- Department of Pharmacology, School of Medicine, University of Patras, Rio Achaias, TK, Greece
| |
Collapse
|
35
|
Cuesta Torres LF, Zhu W, Öhrling G, Larsson R, Patel M, Wiese CB, Rye KA, Vickers KC, Tabet F. High-density lipoproteins induce miR-223-3p biogenesis and export from myeloid cells: Role of scavenger receptor BI-mediated lipid transfer. Atherosclerosis 2019; 286:20-29. [PMID: 31096070 DOI: 10.1016/j.atherosclerosis.2019.04.227] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 04/17/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS We recently showed that miR-223-3p on high-density lipoproteins (HDL) is exported to endothelial cells, where it inhibits inflammation. However, the origin of miR-223-3p on HDL is unknown. We hypothesize that HDL-associated miR-223-3p originates in myeloid cells and is exported to HDL in a scavenger receptor BI (SR-BI)-dependent manner. METHODS Polymorphonuclear neutrophils (PMNs) and human monocyte derived macrophages (HMDMs) were incubated with native HDL (nHDL) or discoidal reconstituted HDL (rHDL). Total RNA was isolated before and after incubation. Mature and primary miR-223-3p (pri-mir-223-3p) levels were quantified by real-time PCR. RESULTS Incubation with nHDL and rHDL increased miR-223-3p export from PMNs and HMDMs. In PMNs, nHDL but not rHDL, increased mature and pri-mir-223-3p. Incubation with HDL also increased Dicer mRNA, a critical regulator of miRNA biogenesis. Incubation of HMDMs with nHDL did not increase cellular levels of mature miR-223-3p, but significantly increased pri-mir-223 levels. Incubation with rHDL had no effect on either mature or pri-mir-223-3p levels. Activated PMNs increased miR-223-3p export to HDL and the production of reactive oxygen species and activated protein kinase C. Blocking HDL binding to SR-BI increased miR-223-3p export to HDL in both PMNs and HMDMs, but did not affect mature and primary miR-223-3p levels. Chemical inhibition of cholesterol flux by Block Lipid Transport (BLT)-1 inhibited HDL-induced pri-mir-223 expression in PMNs. CONCLUSIONS HDL-associated miR-223-3p originates in PMNs and macrophages. HDL stimulates miR-223-3p biogenesis in PMNs in a process that is regulated by SR-BI-mediated lipid flux.
Collapse
Affiliation(s)
| | - Wanying Zhu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gustav Öhrling
- School of Medical Sciences, University of New South Wales Sydney, NSW, Australia
| | - Rasmus Larsson
- School of Medical Sciences, University of New South Wales Sydney, NSW, Australia
| | - Mili Patel
- School of Medical Sciences, University of New South Wales Sydney, NSW, Australia
| | - Carrie B Wiese
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kerry-Anne Rye
- School of Medical Sciences, University of New South Wales Sydney, NSW, Australia
| | - Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Fatiha Tabet
- School of Medical Sciences, University of New South Wales Sydney, NSW, Australia.
| |
Collapse
|
36
|
Huang L, Chambliss KL, Gao X, Yuhanna IS, Behling-Kelly E, Bergaya S, Ahmed M, Michaely P, Luby-Phelps K, Darehshouri A, Xu L, Fisher EA, Ge WP, Mineo C, Shaul PW. SR-B1 drives endothelial cell LDL transcytosis via DOCK4 to promote atherosclerosis. Nature 2019; 569:565-569. [PMID: 31019307 PMCID: PMC6631346 DOI: 10.1038/s41586-019-1140-4] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 03/25/2019] [Indexed: 01/15/2023]
Abstract
Atherosclerosis, which underlies life-threatening cardiovascular disorders including myocardial infarction and stroke1, is initiated by low density lipoprotein cholesterol (LDL) passage into the artery wall and engulfment by macrophages, leading to foam cell formation and lesion development2, 2, 3, 3. How circulating LDL enters the artery wall to instigate atherosclerosis is unknown. Here we show in mice that scavenger receptor, class B type 1 (SR-B1) in endothelial cells mediates LDL delivery into arteries and its accumulation by artery wall macrophages, thereby promoting atherosclerosis. LDL particles are colocalized with SR-B1 in endothelial cell intracellular vesicles in vivo, and LDL transcytosis across endothelial monolayers requires its direct binding to SR-B1 and an 8 amino acid cytoplasmic domain of the receptor that recruits the guanine nucleotide exchange factor dedicator of cytokinesis 4 (DOCK4)4. DOCK4 promotes SR-B1 internalization and LDL transport by coupling LDL binding to SR-B1 with Rac1 activation. SR-B1 and DOCK4 expression are increased in atherosclerosis-prone regions of the mouse aorta prior to lesion formation, and in human atherosclerotic versus normal arteries. These findings challenge the long-held concept that atherogenesis involves passive LDL movement across a compromised endothelial barrier. Interventions inhibiting endothelial delivery of LDL into the artery wall may represent a new therapeutic category in the battle against cardiovascular disease.
Collapse
Affiliation(s)
- Linzhang Huang
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ken L Chambliss
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaofei Gao
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ivan S Yuhanna
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Erica Behling-Kelly
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Sonia Bergaya
- Department of Medicine, New York University School of Medicine, New York, NY, USA.,Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY, USA.,Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, USA
| | - Mohamed Ahmed
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Peter Michaely
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kate Luby-Phelps
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anza Darehshouri
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward A Fisher
- Department of Medicine, New York University School of Medicine, New York, NY, USA.,Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY, USA.,Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, USA
| | - Woo-Ping Ge
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
37
|
Sahebi R, Hassanian SM, Ghayour‐Mobarhan M, Farrokhi E, Rezayi M, Samadi S, Bahramian S, Ferns GA, Avan A. Scavenger receptor Class B type I as a potential risk stratification biomarker and therapeutic target in cardiovascular disease. J Cell Physiol 2019; 234:16925-16932. [DOI: 10.1002/jcp.28393] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Reza Sahebi
- Department of Modern Sciences and Technologies, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Department of Molecular Medicine, School of Advanced Technologies Shahrekord University of Medical Sciences Shahrekord Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Majid Ghayour‐Mobarhan
- Department of Modern Sciences and Technologies, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Effat Farrokhi
- Department of Molecular Medicine, School of Advanced Technologies Shahrekord University of Medical Sciences Shahrekord Iran
| | - Majid Rezayi
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Sara Samadi
- Department of Modern Sciences and Technologies, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Shabbou Bahramian
- Stem Cell Research Center Golestan University of Medical Sciences Gorgan Iran
| | - Gordon A. Ferns
- Division of Medical Education Brighton & Sussex Medical School, Falmer Brighton Sussex
| | - Amir Avan
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
38
|
Abstract
Metabolism and Function of High-Density Lipoproteins (HDL) Abstract. HDL has long been considered as 'good cholesterol', beneficial to the whole body and in particular to cardio-vascular health. However, HDL is a complex particle that undergoes dynamic remodeling through interactions with various enzymes and tissue types throughout its life cycle. In this review, we explore the novel understanding of HDL as a multifaceted class of lipoprotein, with multiple subclasses of different size, molecular composition, receptor interactions, and functionality, in health and disease. Further, we report on emergent HDL based therapeutics tested in small and larger scale clinical trials and their mixed successes.
Collapse
Affiliation(s)
- Anne Jomard
- 1 Eidgenössische Technische Hochschule (ETH), Labor für Translationale Ernährungsbiologie, Zürich
| | - Elena Osto
- 1 Eidgenössische Technische Hochschule (ETH), Labor für Translationale Ernährungsbiologie, Zürich
| |
Collapse
|
39
|
|
40
|
Muresan XM, Sticozzi C, Belmonte G, Cervellati F, Ferrara F, Lila MA, Valacchi G. SR-B1 involvement in keratinocytes in vitro wound closure. Arch Biochem Biophys 2018; 658:1-6. [PMID: 30240595 DOI: 10.1016/j.abb.2018.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 12/17/2022]
Abstract
Skin represents the most extended organ of human body, having as main function the protection of our body from outdoor stressors. Its protective ability is compromised when the skin is disrupted as a consequence of mechanical insults. For this purpose, cutaneous tissue is equipped with an efficient and fine mechanism involved in repairing the wounded area. Among the numerous players that take part in the wound healing process, SR-B1 has been recently shown to have a role in keratinocyte re-epithelialization. SR-B1 is a mediator of cholesterol uptake from HDLs, whereas it is implicated in other cellular processes such as vitamins absorption, vesicle trafficking or pathogen identification. The aim of this study was to investigate the mechanisms involved in SR-B1 role in skin wound closure. Our in vitro data demonstrated that SR-B1 influenced keratinocyte proliferation and migration through a downregulation of nuclear cyclin D1 levels and active MMP9 expression respectively possibly in an NF-kB-dependent mechanism. In addition, SR-B1 was also able to modulate keratinocyte morphology into a pro-migratory cytoskeleton rearrangement. The present in vitro study suggests a new role of SRB1 as a possible new key player in cutaneous wound healing mechanism.
Collapse
Affiliation(s)
- Ximena M Muresan
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Claudia Sticozzi
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Belmonte
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesca Ferrara
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mary Ann Lila
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA
| | - Giuseppe Valacchi
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA.
| |
Collapse
|
41
|
Orekhov AN, Pushkarsky T, Oishi Y, Nikiforov NG, Zhelankin AV, Dubrovsky L, Makeev VJ, Foxx K, Jin X, Kruth HS, Sobenin IA, Sukhorukov VN, Zakiev ER, Kontush A, Le Goff W, Bukrinsky M. HDL activates expression of genes stimulating cholesterol efflux in human monocyte-derived macrophages. Exp Mol Pathol 2018; 105:202-207. [PMID: 30118702 DOI: 10.1016/j.yexmp.2018.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/24/2022]
Abstract
High density lipoproteins (HDL) are key components of reverse cholesterol transport pathway. HDL removes excessive cholesterol from peripheral cells, including macrophages, providing protection from cholesterol accumulation and conversion into foam cells, which is a key event in pathogenesis of atherosclerosis. The mechanism of cellular cholesterol efflux stimulation by HDL involves interaction with the ABCA1 lipid transporter and ensuing transfer of cholesterol to HDL particles. In this study, we looked for additional proteins contributing to HDL-dependent cholesterol efflux. Using RNAseq, we analyzed mRNAs induced by HDL in human monocyte-derived macrophages and identified three genes, fatty acid desaturase 1 (FADS1), insulin induced gene 1 (INSIG1), and the low-density lipoprotein receptor (LDLR), expression of which was significantly upregulated by HDL. We individually knocked down these genes in THP-1 cells using gene silencing by siRNA, and measured cellular cholesterol efflux to HDL. Knock down of FADS1 did not significantly change cholesterol efflux (p = 0.70), but knockdown of INSIG1 and LDLR resulted in highly significant reduction of the efflux to HDL (67% and 75% of control, respectively, p < 0.001). Importantly, the suppression of cholesterol efflux was independent of known effects of these genes on cellular cholesterol content, as cells were loaded with cholesterol using acetylated LDL. These results indicate that HDL particles stimulate expression of genes that enhance cellular cholesterol transfer to HDL.
Collapse
Affiliation(s)
- Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Tatiana Pushkarsky
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Yumiko Oishi
- Department of Cellular and Molecular Medicine, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nikita G Nikiforov
- Institute of General Pathology and Pathophysiology, Moscow, Russia; Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow, Russia; Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey V Zhelankin
- Laboratory of postgenomic research, Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia
| | - Larisa Dubrovsky
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Vsevolod J Makeev
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia; Scientific Center "Kurchatov Institute", Research Institute for Genetics and Selection of Industrial Microorganisms, Moscow, Russia; Moscow Institute of Physics and Technology (State University), Dolgoprudny, Moscow, Region, Russia; Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Kathy Foxx
- Kalen Biomedical LLC, Montgomery Village, MD, USA
| | - Xueting Jin
- Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Howard S Kruth
- Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Igor A Sobenin
- Institute of General Pathology and Pathophysiology, Moscow, Russia; Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow, Russia
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, Moscow, Russia; Sorbonne Université, Inserm, Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, Hôpital de la Pitié, Paris, France
| | - Emile R Zakiev
- Institute of General Pathology and Pathophysiology, Moscow, Russia; Sorbonne Université, Inserm, Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, Hôpital de la Pitié, Paris, France
| | - Anatol Kontush
- Sorbonne Université, Inserm, Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, Hôpital de la Pitié, Paris, France
| | - Wilfried Le Goff
- Sorbonne Université, Inserm, Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, Hôpital de la Pitié, Paris, France
| | - Michael Bukrinsky
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
42
|
Jaafar F, Abdullah A, Makpol S. Cellular Uptake and Bioavailability of Tocotrienol-Rich Fraction in SIRT1-Inhibited Human Diploid Fibroblasts. Sci Rep 2018; 8:10471. [PMID: 29992988 PMCID: PMC6041292 DOI: 10.1038/s41598-018-28708-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 06/28/2018] [Indexed: 02/08/2023] Open
Abstract
Tocotrienol-rich fraction (TRF) is palm vitamin E that consists of tocopherol and tocotrienol. TRF is involved in important cellular regulation including delaying cellular senescence. A key regulator of cellular senescence, Sirtuin 1 (SIRT1) is involved in lipid metabolism. Thus, SIRT1 may regulate vitamin E transportation and bioavailability at cellular level. This study aimed to determine the role of SIRT1 on cellular uptake and bioavailability of TRF in human diploid fibroblasts (HDFs). SIRT1 gene in young HDFs was silenced by small interference RNA (siRNA) while SIRT1 activity was inhibited by sirtinol. TRF treatment was given for 24 h before or after SIRT1 inhibition. Cellular concentration of TRF isomers was determined according to the time points of before and after TRF treatment at 0, 24, 48, 72 and 96 h. Our results showed that all tocotrienol isomers were significantly taken up by HDFs after 24 h of TRF treatment and decreased 24 h after TRF treatment was terminated but remained in the cell up to 72 h. The uptake of α-tocopherol, α-tocotrienol and β-tocotrienol was significantly higher in senescent cells as compared to young HDFs indicating higher requirement for vitamin E in senescent cells. Inhibition of SIRT1 gene increased the uptake of all tocotrienol isomers but not α-tocopherol. However, SIRT1 inhibition at protein level decreased tocotrienol concentration. In conclusion, SIRT1 may regulate the cellular uptake and bioavailability of tocotrienol isomers in human diploid fibroblast cells while a similar regulation was not shown for α-tocopherol.
Collapse
Affiliation(s)
- Faizul Jaafar
- Department of Biochemistry, Faculty of Medicine, Level 17, Preclinical Building, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Asmaa Abdullah
- Department of Biochemistry, Faculty of Medicine, Level 17, Preclinical Building, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Level 17, Preclinical Building, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
43
|
Cifarelli V, Abumrad NA. Intestinal CD36 and Other Key Proteins of Lipid Utilization: Role in Absorption and Gut Homeostasis. Compr Physiol 2018; 8:493-507. [PMID: 29687890 PMCID: PMC6247794 DOI: 10.1002/cphy.c170026] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Several proteins have been implicated in fatty acid (FA) transport by enterocytes including the scavenger receptor CD36 (SR-B2), the scavenger receptor B1 (SR-B1) a member of the CD36 family and the FA transport protein 4 (FATP4). Here, we review the regulation of enterocyte FA uptake and its function in lipid absorption including prechylomicron formation, assembly and transport. Emphasis is given to CD36, which is abundantly expressed along the digestive tract of rodents and humans and has been the most studied. We also address the pleiotropic functions of CD36 that go beyond lipid absorption and metabolism to include recent evidence of its impact on intestinal homeostasis and barrier maintenance. Areas of progress involving contribution of membrane phospholipid remodeling and of cytosolic FA-binding proteins, FABP1 and FABP2 to fat absorption will be covered. © 2018 American Physiological Society. Compr Physiol 8:493-507, 2018.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Internal Medicine, Center for Human Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| | - Nada A. Abumrad
- Department of Internal Medicine, Center for Human Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
44
|
Singh PK, Jaiswal AK, Pawar VK, Raval K, Kumar A, Bora HK, Dube A, Chourasia MK. Fabrication of 3-O-sn-Phosphatidyl-L-serine Anchored PLGA Nanoparticle Bearing Amphotericin B for Macrophage Targeting. Pharm Res 2018; 35:60. [DOI: 10.1007/s11095-017-2293-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/24/2017] [Indexed: 12/22/2022]
|
45
|
de Lima AD, Guido MC, Tavares ER, Carvalho PO, Marques AF, de Melo MDT, Salemi VMC, Kalil-Filho R, Maranhão RC. The Expression of Lipoprotein Receptors Is Increased in the Infarcted Area After Myocardial Infarction Induced in Rats With Cardiac Dysfunction. Lipids 2018; 53:177-187. [DOI: 10.1002/lipd.12014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Aline D. de Lima
- Laboratory of Metabolism and Lipids; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| | - Maria C. Guido
- Laboratory of Metabolism and Lipids; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| | - Elaine R. Tavares
- Laboratory of Metabolism and Lipids; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| | - Priscila O. Carvalho
- Laboratory of Metabolism and Lipids; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| | - Alyne F. Marques
- Laboratory of Metabolism and Lipids; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| | - Marcelo D. T. de Melo
- Heart Failure Unit and Clinical Cardiology Division; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| | - Vera M. C. Salemi
- Heart Failure Unit and Clinical Cardiology Division; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| | - Roberto Kalil-Filho
- Heart Failure Unit and Clinical Cardiology Division; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| | - Raul C. Maranhão
- Laboratory of Metabolism and Lipids; Heart Institute (InCor), Medical School Hospital, University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
- Faculty of Pharmaceutical Sciences; University of São Paulo; Av. Dr. Eneas de Carvalho Aguiar, 44, Bl. 2, 1o.SS, São Paulo SP, 05403-900 Brazil
| |
Collapse
|
46
|
de Boer JF, Kuipers F, Groen AK. Cholesterol Transport Revisited: A New Turbo Mechanism to Drive Cholesterol Excretion. Trends Endocrinol Metab 2018; 29:123-133. [PMID: 29276134 DOI: 10.1016/j.tem.2017.11.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 11/26/2017] [Accepted: 11/29/2017] [Indexed: 12/31/2022]
Abstract
A fine-tuned balance between cholesterol uptake and excretion by the body is pivotal to maintain health and to remain free from the deleterious consequences of cholesterol accumulation such as cardiovascular disease. The pathways involved in intracellular and extracellular cholesterol transport are a subject of intense investigation and are being unraveled in increasing detail. In addition, insight into the complex interactions between cholesterol and bile acid metabolism has increased considerably in the last couple of years. This review provides an overview of the mechanisms involved in cholesterol uptake and excretion, with a particular emphasis on the most recent progress in this field. Special attention is given to the transintestinal cholesterol excretion (TICE) pathway, which was recently demonstrated to have a remarkably high transport capacity and to be sensitive to pharmacological modulation.
Collapse
Affiliation(s)
- Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert K Groen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Vascular Medicine, University of Amsterdam Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Muresan XM, Sticozzi C, Belmonte G, Savelli V, Evelson P, Valacchi G. Modulation of cutaneous scavenger receptor B1 levels by exogenous stressors impairs "in vitro" wound closure. Mech Ageing Dev 2017; 172:78-85. [PMID: 29102450 DOI: 10.1016/j.mad.2017.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/30/2017] [Accepted: 11/01/2017] [Indexed: 01/01/2023]
Abstract
Scavenger receptor B1 (SR-B1) is a trans-membrane protein, involved in tissue reverse cholesterol transport. Several studies have demonstrated that SR-B1 is also implicated in other physiological processes, such as bacteria and apoptotic cells recognition and regulation of intracellular tocopherol and carotenoids levels. Among the tissues where it is localized, SR-B1 has been shown to be significantly expressed in human epidermis. Our group has demonstrated that SR-B1 levels are down-regulated in human cultured keratinocytes by environmental stressors, such as cigarette smoke, via cellular redox imbalance. Our present study aimed to investigate whether such down-regulation was confirmed in a 3D skin model and under other environmental challengers such as particulate matter and ozone. We also investigated the association between oxidation-induced SR-B1 modulation and impaired wound closure. The data obtained showed that not only cigarette, but also the other environmental stressors reduced SR-B1 expression in epidermal cutaneous tissues and that this effect might be involved in impaired wound healing.
Collapse
Affiliation(s)
| | - Claudia Sticozzi
- Dept. of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Belmonte
- Dept. of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Vinno Savelli
- Department of Medical, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Pablo Evelson
- Institute of Biochemistry and Molecular Medicine (IBIMOL-UBA-CONICET), Pharmacy and Biochemistry School, University of Buenos Aires, Buenos Aires, Argentina
| | - Giuseppe Valacchi
- Dept. of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA.
| |
Collapse
|
48
|
Kato T, Lee D, Huang H, Cruz W, Ujiie H, Fujino K, Wada H, Patel P, Hu HP, Hirohashi K, Nakajima T, Sato M, Kaji M, Kaga K, Matsui Y, Chen J, Zheng G, Yasufuku K. Personalized siRNA-Nanoparticle Systemic Therapy using Metastatic Lymph Node Specimens Obtained with EBUS-TBNA in Lung Cancer. Mol Cancer Res 2017; 16:47-57. [PMID: 28993508 DOI: 10.1158/1541-7786.mcr-16-0341] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/25/2017] [Accepted: 10/04/2017] [Indexed: 11/16/2022]
Abstract
Inhibiting specific gene expression with siRNA provides a new therapeutic strategy to tackle many diseases at the molecular level. Recent strategies called high-density lipoprotein (HDL)-mimicking peptide-phospholipid nanoscaffold (HPPS) nanoparticles have been used to induce siRNAs-targeted delivery to scavenger receptor class B type I receptor (SCARB1)-expressing cancer cells with high efficiency. Here, eight ideal therapeutic target genes were identified for advanced lung cancer throughout the screenings using endobronchial ultrasonography-guided transbronchial needle aspiration (EBUS-TBNA) and the establishment of a personalized siRNA-nanoparticle therapy. The relevance of these genes was evaluated by means of siRNA experiments in cancer cell growth. To establish a therapeutic model, kinesin family member-11 (KIF11) was selected as a target gene. A total of 356 lung cancers were analyzed immunohistochemically for its clinicopathologic significance. The antitumor effect of HPPS-conjugated siRNA was evaluated in vivo using xenograft tumor models. Inhibition of gene expression for these targets effectively suppressed lung cancer cell growth. SCARB1 was highly expressed in a subset of tumors from the lung large-cell carcinoma (LCC) and small-cell lung cancer (SCLC) patients. High-level KIF11 expression was identified as an independent prognostic factor in LCC and squamous cell carcinoma (SqCC) patients. Finally, a conjugate of siRNA against KIF11 and HPPS nanoparticles induced downregulation of KIF11 expression and mediated dramatic inhibition of tumor growth in vivoImplications: This approach showed delivering personalized cancer-specific siRNAs via the appropriate nanocarrier may be a novel therapeutic option for patients with advanced lung cancer. Mol Cancer Res; 16(1); 47-57. ©2017 AACR.
Collapse
Affiliation(s)
- Tatsuya Kato
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.,Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Daiyoon Lee
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Huang Huang
- DLVR Therapeutics Inc. and University Health Network, Toronto, Canada
| | - William Cruz
- DLVR Therapeutics Inc. and University Health Network, Toronto, Canada
| | - Hideki Ujiie
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Kosuke Fujino
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Hironobu Wada
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.,Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Chiba, Japan
| | - Priya Patel
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Hsin-Pei Hu
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Kentaro Hirohashi
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Takahiro Nakajima
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Chiba, Japan
| | - Masaaki Sato
- Department of Pathology, NTT East Japan Sapporo Hospital, Sapporo, Hokkaido Japan
| | - Mitsuhito Kaji
- Department of Thoracic Surgery, Sapporo Minami-Sanjo Hospital, Sapporo, Hokkaido, Japan
| | - Kichizo Kaga
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Yoshiro Matsui
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Juan Chen
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Gang Zheng
- DLVR Therapeutics Inc. and University Health Network, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Kazuhiro Yasufuku
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
49
|
Hu G, He M, Ko WKW, Wong AOL. TAC1 Gene Products Regulate Pituitary Hormone Secretion and Gene Expression in Prepubertal Grass Carp Pituitary Cells. Endocrinology 2017; 158:1776-1797. [PMID: 28323939 DOI: 10.1210/en.2016-1740] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 03/07/2017] [Indexed: 01/30/2023]
Abstract
Tachykinin-1 (TAC1) is known to have diverse functions in mammals, but similar information is scarce in fish species. Using grass carp as a model, the pituitary actions, receptor specificity and postreceptor signaling of TAC1 gene products, namely substance P (SP) and neurokinin A (NKA), were examined. TAC1 encoding SP and NKA as well as tachykinin receptors NK1R and NK2R were cloned in the carp pituitary. The newly cloned receptors were shown to be functional with properties similar to mammalian counterparts. In carp pituitary cells, SP and NKA could trigger luteinizing hormone (LH), prolactin (PRL), and somatolactin α (SLα) secretion, with parallel rises in PRL and SLα transcripts. Short-term SP treatment (3 hours) induced LH release, whereas prolonged induction (24 hours) could attenuate LHβ messenger RNA (mRNA) expression. At pituitary cell level, LH, PRL, and SLα regulation by TAC1 gene products were mediated by NK1R, NK2R, and NK3R, respectively. Apparently, SP- and NKA-induced LH and SLα secretion and transcript expression were mediated by adenylyl cyclase/cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA), phospholiphase C (PLC)/inositol 1,4,5-triphosphate/protein kinase C (PKC), and Ca2+/calmodulin (CaM)/CaM-dependent protein kinase-II pathways. The signal transduction for PRL responses was similar, except for the absence of a PKC component. Regarding SP inhibition of LHβ mRNA expression, the cAMP/PKA- and PLC/PKC-dependent (but not Ca2+/CaM-dependent) cascades were involved. These results, as a whole, suggest that TAC1 gene products play a role in LH, PRL, and SLα regulation via overlapping postreceptor signaling coupled to different subtypes of tachykinin receptor expressed in the carp pituitary.
Collapse
Affiliation(s)
- Guangfu Hu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Mulan He
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Wendy K W Ko
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Anderson O L Wong
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| |
Collapse
|
50
|
Regulated Entry of Hepatitis C Virus into Hepatocytes. Viruses 2017; 9:v9050100. [PMID: 28486435 PMCID: PMC5454413 DOI: 10.3390/v9050100] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/24/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) is a model for the study of virus–host interaction and host cell responses to infection. Virus entry into hepatocytes is the first step in the HCV life cycle, and this process requires multiple receptors working together. The scavenger receptor class B type I (SR-BI) and claudin-1 (CLDN1), together with human cluster of differentiation (CD) 81 and occludin (OCLN), constitute the minimal set of HCV entry receptors. Nevertheless, HCV entry is a complex process involving multiple host signaling pathways that form a systematic regulatory network; this network is centrally controlled by upstream regulators epidermal growth factor receptor (EGFR) and transforming growth factor β receptor (TGFβ-R). Further feedback regulation and cell-to-cell spread of the virus contribute to the chronic maintenance of HCV infection. A comprehensive and accurate disclosure of this critical process should provide insights into the viral entry mechanism, and offer new strategies for treatment regimens and targets for HCV therapeutics.
Collapse
|