51
|
Zhang D, Zhang R, Liu Y, Sun X, Yin Z, Li H, Zhao Y, Wang B, Ren Y, Cheng C, Liu X, Liu D, Liu F, Chen X, Liu L, Zhou Q, Xiong Y, Xu Q, Liu J, Hong S, You Z, Hu D, Zhang M. CD36 gene variants is associated with type 2 diabetes mellitus through the interaction of obesity in rural Chinese adults. Gene 2018; 659:155-159. [PMID: 29572193 DOI: 10.1016/j.gene.2018.03.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/14/2018] [Accepted: 03/19/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Evidences show that cluster determinant 36 (CD36) protein plays a role in lipid metabolism and insulin resistance, and the expression of CD36 is inducible in obesity. The present study evaluated the association of CD36 variants and the interaction with obesity on type 2 diabetes mellitus (T2DM) risk. METHODS We performed a case-control study nested in the Rural Chinese Cohort Study. We included 546 incident T2DM cases matched with non-T2DM controls in a 1:1 ratio by sex, age (within 2 years), marital status, and residence village. Four loci in CD36 (rs1194197, rs2151916, rs3211956, and rs7755) were genotyped by SNPscanTM Genotyping system. RESULTS After adjusting for potential confounding, we observed no statistically significant association between the CD36 polymorphisms and T2DM risk. Compared to wild-type homozygous carriers with normal weight, overweight/obesity participants carrying the mutational allele rs7755 showed increased risk of T2DM, by 114% (OR = 2.14, 95% CI: 1.33-3.46; Pinteraction = 0.007); abdominal obesity participants carrying the mutational allele rs7755 showed increased risk of T2DM, by 133% (OR = 2.33, 95% CI: 1.48-3.66; Pinteraction = 0.002). Furthermore, rs2151916 polymorphism was associated with triglycerides level (P = 0.019), and the rs1194197 variant was related to systolic blood pressure (P = 0.023) within the group of controls. CONCLUSIONS CD36 genotypes were not associated with the progression to T2DM independently. However, our results suggested a positive interaction between the CD36 variants and obesity on T2DM susceptibility, which might be through a cardiometabolic disorder.
Collapse
Affiliation(s)
- Dongdong Zhang
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Ruiyuan Zhang
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yu Liu
- The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Xizhuo Sun
- The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Zhaoxia Yin
- The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Honghui Li
- The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yang Zhao
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Bingyuan Wang
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yongcheng Ren
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Cheng Cheng
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Xuejiao Liu
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Dechen Liu
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Feiyan Liu
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Xu Chen
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Leilei Liu
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Qionggui Zhou
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yihan Xiong
- Department of Clinical Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Qihuan Xu
- Department of Clinical Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Jiali Liu
- Department of Clinical Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Shihao Hong
- Department of Clinical Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Ziyang You
- Department of Clinical Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Dongsheng Hu
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Ming Zhang
- Department of Preventive Medicine, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China; Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
52
|
Lack of pronounced changes in the expression of fatty acid handling proteins in adipose tissue and plasma of morbidly obese humans. Nutr Diabetes 2018; 8:3. [PMID: 29335416 PMCID: PMC5851429 DOI: 10.1038/s41387-017-0013-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 10/20/2017] [Accepted: 11/17/2017] [Indexed: 12/11/2022] Open
Abstract
Background/Objectives Fatty acid handling proteins are involved in the process of accumulation of lipids in different fat tissue depots. Thus, the aim of the study was to estimate the expression of both fatty acid transport and binding proteins in the subcutaneous (SAT) and visceral adipose tissue (VAT) of patients with morbid obesity without metabolic syndrome, as well as the plasma concentrations of these transporters. Subjects/Methods Protein (Western blotting) and mRNA (Real-time PCR) expression of selected fatty acid handling proteins was assessed in the visceral and subcutaneous adipose tissue of 30 patients with morbid obesity. The control group consisted of 10 lean age-matched patients. Plasma levels of fatty acid protein transporters were also evaluated using ELISA method. Moreover, total plasma fatty acid composition and concentration was determined by gas-liquid chromatography (GLC). Results Significant increase in fatty acid translocase (FAT/CD36) mRNA (P = 0.03) and plasmalemmal (P = 0.01) expression was observed in VAT of patients with morbid obesity vs. lean subjects together with elevation of lipoprotein lipase (LPL), as well as peroxisome proliferator-activated receptor γ (PPARγ) in both examined compartments of adipose tissue. Moreover, in obese subjects plasma concentration of RBP4 was markedly elevated (P = 0.04) and sCD36 level presented a tendency for an increase (P = 0.08) with concomitant lack of changes in FABP4 concentration (P > 0.05). Conclusions Fatty acid transport into adipocytes may be, at least in part, related to the increased expression of FAT/CD36 in the VAT of morbidly obese patients, which is accompanied by augmented expression of LPL, as well as PPARγ. Probably, alternations in plasma concentrations of RBP4 and sCD36 in obese patients are associated with “unhealthy” fat distribution.
Collapse
|
53
|
CD36 in chronic kidney disease: novel insights and therapeutic opportunities. Nat Rev Nephrol 2017; 13:769-781. [DOI: 10.1038/nrneph.2017.126] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
54
|
Chu Y, Lao W, Jin G, Dai D, Chen L, Kang H. Evaluation of the relationship between CD36 and MARCO single-nucleotide polymorphisms and susceptibility to carotid atherosclerosis in a Chinese Han population. Gene 2017; 633:66-70. [PMID: 28866086 DOI: 10.1016/j.gene.2017.08.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/29/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE This study analyzed the genetic association between two scavenger receptors single nucleotide polymorphisms (CD36 rs1761667, MARCO rs12998782) and carotid atherosclerosis in a Chinese Han population. METHODS Samples of genomic DNA collected from patients (n=215) and healthy control subjects (n=252) were analyzed by the polymerase chain reaction with high-resolution melting analysis. Odds ratios and 95% confidence intervals were used to evaluate the association between the two SNPs and carotid atherosclerosis. RESULTS There was no difference between the SNPs regarding their association with the frequency of carotid atherosclerosis in the case and control groups or in the male case group and control group. Female patients of genotype GA for CD36 rs1761667 and CT for MARCO rs12998782 were at an increased risk for carotid atherosclerosis. The presence of rs1761667 GA and rs12998782 CT may increase the risk for carotid atherosclerosis among postmenopausal females. CONCLUSIONS CD36 and MARCO are associated with the susceptibility of Chinese Han females to carotid atherosclerosis. Menopausal status may affect the association between gene polymorphisms and carotid atherosclerosis in the female Chinese Han population.
Collapse
Affiliation(s)
- Yang Chu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Wenting Lao
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Guojiang Jin
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Di Dai
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Li Chen
- Department of Blood Transfusion, Shanghai East Hospital, Tongji University, Shanghai 200120, China
| | - Hui Kang
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China.
| |
Collapse
|
55
|
Kim HJ, Moon JS, Park IR, Kim JH, Yoon JS, Won KC, Lee HW. A Novel Index Using Soluble CD36 Is Associated with the Prevalence of Type 2 Diabetes Mellitus: Comparison Study with Triglyceride-Glucose Index. Endocrinol Metab (Seoul) 2017; 32:375-382. [PMID: 28956368 PMCID: PMC5620035 DOI: 10.3803/enm.2017.32.3.375] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Plasma soluble cluster determinant 36 (sCD36) level is closely related with insulin resistance and atherosclerosis, but little is known whether it could be a surrogate for estimating risk of developing diabetes or not. To address this, we evaluated association between sCD36 index, the product of sCD36 and fasting plasma glucose (FPG), and the prevalence of type 2 diabetes mellitus (T2DM), and then compared with triglyceride-glucose (TyG) index which has been suggested simple index for insulin resistance. METHODS This was cross-sectional study, and participants were classified as normal glucose tolerance (NGT), prediabetes, and T2DM according to glucose tolerance. The formula of TyG index was 'ln [FPG (mg/dL)×triglyceride (mg/dL)/2],' and the sCD36 index was 'ln [sCD36 (pg/mL)×FPG (mg/dL)/2].' RESULTS One hundred and fifty-five subjects (mean age, 55.2 years) were enrolled, and patients with T2DM were 75. Both indexes were significantly increased in prediabetes and T2DM rather than NGT, and sCD36 index was positively correlated with both glycosylated hemoglobin and homeostasis model assessment of insulin resistance (r=0.767 and r=0.453, respectively; P<0.05) and negatively with homeostasis model assessment estimate of β-cell function (r=-0.317). The odds ratio (OR) of sCD36 index for T2DM was 4.39 (95% confidential interval, 1.51 to 12.77) after adjusting age, gender, blood pressure, smoking, alcohol, non-high density lipoprotein cholesterol and high-sensitivity C-reactive protein. However, OR of TyG index did not remained significance after adjustment. CONCLUSION sCD36 index has an independent association with the risk of T2DM, and showed better correlation than TyG index. These results suggest sCD36 index might be useful surrogate marker for the risk of diabetes.
Collapse
Affiliation(s)
- Ho Jin Kim
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jun Sung Moon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Il Rae Park
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Joong Hee Kim
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Ji Sung Yoon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyu Chang Won
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Hyoung Woo Lee
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea.
| |
Collapse
|
56
|
Lopez-Carmona MD, Plaza-Seron MC, Vargas-Candela A, Tinahones FJ, Gomez-Huelgas R, Bernal-Lopez MR. CD36 overexpression: a possible etiopathogenic mechanism of atherosclerosis in patients with prediabetes and diabetes. Diabetol Metab Syndr 2017; 9:55. [PMID: 28729885 PMCID: PMC5516302 DOI: 10.1186/s13098-017-0253-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 07/12/2017] [Indexed: 01/28/2023] Open
Abstract
RATIONALE CD36 is a scavenger receptor located on monocytes which is involved in foam cell transformation. AIM To evaluate CD36 expression under different glycemic states in both healthy subjects and in atherosclerotic patients. SUBJECTS AND METHODS In order to evaluate the possible effects of hyperglycemia on CD36 expression in healthy subjects, an in vitro experiment was carried out using monocyte in three different conditions: extreme hyperglycemia (HG), euglycemia (EG) and in the absence of glucose. On the other hand, three groups of atherosclerotic patients were evaluated according to their glycemic conditions: normoglycemic (NG), prediabetic (preDM) and diabetic (DM) patients. CD36 expression (mRNA, non-glycated and glycated protein) was analyzed in monocytes. RESULTS CD36 mRNA expression in the in vitro experiment peaked at 4 and 24 h under HG conditions. No differences in mRNA levels were found in the EG and control group. The level of non-glycated proteins was higher in HG and EG conditions compared with control group. Glycated protein expression was inhibited by glucose in a sustained manner. In atherosclerotic patients, a significant association was observed when comparing glycated CD36 protein expression in DM with NG patients (p = 0.03). No significant differences were found in mRNA and non-glycated CD36 expression in these patients. Moreover, BMI, insulin, weight and treatment were shown to be related to CD36 expression (mRNA, non-glycated and glycated protein levels, depending of the case) in atherosclerotic patients. CONCLUSIONS Hyperglycemia is an important modulator of CD36 mRNA and non-glycated protein expression in vitro, increasing de novo synthesis in healthy subjects. In atherosclerotic patients, there are progressive increases in CD36 receptors, which may be due to a post-translational stimulus.
Collapse
Affiliation(s)
- M. D. Lopez-Carmona
- Internal Medicine Department, Biomedical Institute of Malaga (IBIMA), Regional University Hospital of Malaga (Carlos Haya Hospital), Avda. Hospital Civil s/n, 29009 Malaga, Spain
| | - M. C. Plaza-Seron
- Research Laboratory-Allergy Unit, Biomedical Institute of Malaga (IBIMA), Regional University Hospital of Malaga (Carlos Haya Hospital), Malaga, Spain
| | - A. Vargas-Candela
- Internal Medicine Department, Biomedical Institute of Malaga (IBIMA), Regional University Hospital of Malaga (Carlos Haya Hospital), Avda. Hospital Civil s/n, 29009 Malaga, Spain
| | - F. J. Tinahones
- Endocrinology and Nutrition Department, Biomedical Institute of Malaga (IBIMA), Regional University Hospital of Malaga (Virgen de la Victoria Hospital), Malaga, Spain
- CIBERFisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - R. Gomez-Huelgas
- Internal Medicine Department, Biomedical Institute of Malaga (IBIMA), Regional University Hospital of Malaga (Carlos Haya Hospital), Avda. Hospital Civil s/n, 29009 Malaga, Spain
- CIBERFisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - M. R. Bernal-Lopez
- Internal Medicine Department, Biomedical Institute of Malaga (IBIMA), Regional University Hospital of Malaga (Carlos Haya Hospital), Avda. Hospital Civil s/n, 29009 Malaga, Spain
- CIBERFisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
57
|
Botha J, Velling Magnussen L, Nielsen MH, Nielsen TB, Højlund K, Andersen MS, Handberg A. Microvesicles Correlated with Components of Metabolic Syndrome in Men with Type 2 Diabetes Mellitus and Lowered Testosterone Levels But Were Unaltered by Testosterone Therapy. J Diabetes Res 2017; 2017:4257875. [PMID: 28168203 PMCID: PMC5266820 DOI: 10.1155/2017/4257875] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/24/2016] [Accepted: 12/14/2016] [Indexed: 01/15/2023] Open
Abstract
Aims. To investigate how circulating microvesicle phenotypes correlate with insulin sensitivity, body composition, plasma lipids, and hepatic fat accumulation. We hypothesized that changes elicited by testosterone replacement therapy are reflected in levels of microvesicles. Methods. Thirty-nine type 2 diabetic males with lowered testosterone levels were assigned to either testosterone replacement therapy or placebo and evaluated at baseline and after 24 weeks. Microvesicles were analysed by flow cytometry and defined as lactadherin-binding particles within the 0.1-1.0 μm gate. Microvesicles of platelet, monocyte, and endothelial cell origin were identified by cell-specific markers and their expression of CD36 was investigated. Results. Triglycerides correlated positively with all investigated microvesicle phenotypes in this study (p < 0.05), and indicators of hepatic fat accumulation, alanine aminotransferase, and gamma glutamyltransferase correlated with platelet and endothelial microvesicles and CD36-expressing microvesicles from platelets and monocytes (p < 0.05). BMI, waist circumference, and fat percentage correlated with CD36-expressing monocyte microvesicles (p < 0.05), while insulin sensitivity did not correlate with any microvesicle phenotypes. Microvesicle levels were unaffected by testosterone therapy. Conclusions. Metabolic syndrome components and hepatic fat accumulation correlated with microvesicle phenotypes, supporting the involvement of especially CD36 on monocytes in metabolic syndrome pathogenesis. Although testosterone therapy improved body composition measures, microvesicle phenotype levels were unaffected. This trial was registered at ClinicalTrials.gov (NCT01560546).
Collapse
Affiliation(s)
- Jaco Botha
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- *Jaco Botha:
| | | | | | - Tine Bo Nielsen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Kurt Højlund
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
- Section of Molecular Diabetes & Metabolism, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
58
|
Madrigal-Ruíz PM, Navarro-Hernández RE, Ruíz-Quezada SL, Corona-Meraz FI, Vázquez-Del Mercado M, Gómez-Bañuelos E, Castro-Albarran J, Sandoval-García F, Flores-Alvarado LJ, Martín-Marquez BT. Low CD36 and LOX-1 Levels and CD36 Gene Subexpression Are Associated with Metabolic Dysregulation in Older Individuals with Abdominal Obesity. J Diabetes Res 2016; 2016:5678946. [PMID: 27525284 PMCID: PMC4976145 DOI: 10.1155/2016/5678946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/17/2016] [Accepted: 06/19/2016] [Indexed: 12/28/2022] Open
Abstract
Background. Obesity study in the context of scavenger receptors has been linked to atherosclerosis. CD36 and LOX-1 are important, since they have been associated with atherogenic and metabolic disease but not fat redistribution. The aim of our study was to determinate the association between CD36 and LOX-1 in presence of age and abdominal obesity. Methods. This is a cross-sectional study that included 151 healthy individuals, clinically and anthropometrically classified into two groups by age (<30 and ≥30 years old) and abdominal obesity (according to World Health Organization guidelines). We excluded individuals with any chronic and metabolic illness, use of medication, or smoking. Fasting blood samples were taken to perform determination of CD36 mRNA expression by real-time PCR, lipid profile and metabolic and low grade inflammation markers by routine methods, and soluble scavenger receptors (CD36 and LOX-1) by ELISA. Results. Individuals ≥30 years old with abdominal obesity presented high atherogenic index, lower soluble scavenger receptor levels, and subexpression of CD36 mRNA (54% less). On the other hand, individuals <30 years old with abdominal adiposity presented higher levels in the same parameters, except LOX-1 soluble levels. Conclusion. In this study, individuals over 30 years of age presented low soluble scavenger receptors levels pattern and CD36 gene subexpression, which suggest the chronic metabolic dysregulation in abdominal obesity.
Collapse
Affiliation(s)
- Perla-Monserrat Madrigal-Ruíz
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- UDG-CA-701, Grupo de Investigación Inmunometabolismo en Enfermedades Emergentes (GIIEE), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| | - Rosa-Elena Navarro-Hernández
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- UDG-CA-701, Grupo de Investigación Inmunometabolismo en Enfermedades Emergentes (GIIEE), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| | - Sandra-Luz Ruíz-Quezada
- UDG-CA-701, Grupo de Investigación Inmunometabolismo en Enfermedades Emergentes (GIIEE), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Boulevard Marcelino García Barragán No. 1421, 44430 Guadalajara, JAL, Mexico
| | - Fernanda-Isadora Corona-Meraz
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- UDG-CA-701, Grupo de Investigación Inmunometabolismo en Enfermedades Emergentes (GIIEE), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| | - Mónica Vázquez-Del Mercado
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- Servicio de Reumatología, División de Medicina Interna, Hospital Civil “Dr. Juan I. Menchaca”, Universidad de Guadalajara, Salvador de Quevedo y Zubieta No. 750, 44340 Guadalajara, JAL, Mexico
- UDG-CA-703, Grupo de Investigación en Inmunología y Reumatología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| | - Eduardo Gómez-Bañuelos
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- UDG-CA-701, Grupo de Investigación Inmunometabolismo en Enfermedades Emergentes (GIIEE), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- UDG-CA-703, Grupo de Investigación en Inmunología y Reumatología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| | - Jorge Castro-Albarran
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- UDG-CA-701, Grupo de Investigación Inmunometabolismo en Enfermedades Emergentes (GIIEE), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| | - Flavio Sandoval-García
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- Departamento de Clínicas Médicas, Antiguo Hospital Civil de Guadalajara, Calle Hospital No. 320, Colonia El Retiro, 44360 Guadalajara, JAL, Mexico
| | - Luis-Javier Flores-Alvarado
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- UDG-CA-701, Grupo de Investigación Inmunometabolismo en Enfermedades Emergentes (GIIEE), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| | - Beatriz-Teresita Martín-Marquez
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
- UDG-CA-703, Grupo de Investigación en Inmunología y Reumatología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada No. 950, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| |
Collapse
|
59
|
Yu M, Jiang M, Chen Y, Zhang S, Zhang W, Yang X, Li X, Li Y, Duan S, Han J, Duan Y. Inhibition of Macrophage CD36 Expression and Cellular Oxidized Low Density Lipoprotein (oxLDL) Accumulation by Tamoxifen: A PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR (PPAR)γ-DEPENDENT MECHANISM. J Biol Chem 2016; 291:16977-89. [PMID: 27358406 DOI: 10.1074/jbc.m116.740092] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Indexed: 12/22/2022] Open
Abstract
Macrophage CD36 binds and internalizes oxidized low density lipoprotein (oxLDL) to facilitate foam cell formation. CD36 expression is activated by peroxisome proliferator-activated receptor γ (PPARγ). Tamoxifen, an anti-breast cancer medicine, has demonstrated pleiotropic functions including cardioprotection with unfully elucidated mechanisms. In this study, we determined that treatment of ApoE-deficient mice with tamoxifen reduced atherosclerosis, which was associated with decreased CD36 and PPARγ expression in lesion areas. At the cellular level, we observed that tamoxifen inhibited CD36 protein expression in human THP-1 monocytes, THP-1/PMA macrophages, and human blood monocyte-derived macrophages. Associated with decreased CD36 protein expression, tamoxifen reduced cellular oxLDL accumulation in a CD36-dependent manner. At the transcriptional level, tamoxifen decreased CD36 mRNA expression, promoter activity, and the binding of the PPARγ response element in CD36 promoter to PPARγ protein. Tamoxifen blocked ligand-induced PPARγ nuclear translocation and CD36 expression, but it increased PPARγ phosphorylation, which was due to that tamoxifen-activated ERK1/2. Furthermore, deficiency of PPARγ expression in macrophages abolished the inhibitory effect of tamoxifen on CD36 expression or cellular oxLDL accumulation both in vitro and in vivo Taken together, our study demonstrates that tamoxifen inhibits CD36 expression and cellular oxLDL accumulation by inactivating the PPARγ signaling pathway, and the inhibition of macrophage CD36 expression can be attributed to the anti-atherogenic properties of tamoxifen.
Collapse
Affiliation(s)
- Miao Yu
- From the College of Life Sciences
| | - Meixiu Jiang
- the Institute of Translational Medicine, Nanchang University, Nanchang 330000
| | - Yuanli Chen
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230009, and School of Medicine, and
| | | | | | | | | | - Yan Li
- From the College of Life Sciences
| | - Shengzhong Duan
- the Institute for Nutritional Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jihong Han
- From the College of Life Sciences, the College of Biomedical Engineering, Hefei University of Technology, Hefei 230009, and the State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin 300071,
| | - Yajun Duan
- From the College of Life Sciences, the College of Biomedical Engineering, Hefei University of Technology, Hefei 230009, and the State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin 300071,
| |
Collapse
|
60
|
Reduced sCD36 following weight loss corresponds to improved insulin sensitivity, dyslipidemia and liver fat in obese children. Eur J Clin Nutr 2016; 70:1073-7. [PMID: 27273071 DOI: 10.1038/ejcn.2016.88] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/24/2016] [Accepted: 04/24/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND/OBJECTIVES Childhood obesity is a major health problem with serious long-term metabolic consequences. CD36 is important for the development of obesity-related complications among adults. We aimed to investigate circulating sCD36 during weight loss in childhood obesity and its associations with insulin resistance, dyslipidemia, hepatic fat accumulation and low-grade inflammation. SUBJECTS/METHODS The impact of a 10-week weight loss camp for obese children (N=113) on plasma sCD36 and further after a 12-month follow-up (N=68) was investigated. Clinical and biochemical data were collected, and sCD36 was measured by an in-house assay. Liver fat was estimated by ultrasonography and insulin resistance by the homeostasis model assessment (HOMA-IR). RESULTS Along with marked weight loss, sCD36 was reduced by 21% (P=0.0013) following lifestyle intervention, and individual sCD36 reductions were significantly associated with the corresponding decreases in HOMA-IR, triglycerides and total cholesterol. The largest sCD36 decrease occurred among children who reduced HOMA-IR and liver fat. After 12 months of follow-up, sCD36 was increased (P=0.014) and the metabolic improvements were largely lost. CONCLUSIONS Weight-loss-induced sCD36 reduction, coincident with improved insulin resistance, circulating lipids and hepatic fat accumulation, proposes that sCD36 may be an early marker of long-term health risk associated with obesity-related complications.
Collapse
|
61
|
Yang X, Zhang W, Chen Y, Li Y, Sun L, Liu Y, Liu M, Yu M, Li X, Han J, Duan Y. Activation of Peroxisome Proliferator-activated Receptor γ (PPARγ) and CD36 Protein Expression: THE DUAL PATHOPHYSIOLOGICAL ROLES OF PROGESTERONE. J Biol Chem 2016; 291:15108-18. [PMID: 27226602 DOI: 10.1074/jbc.m116.726737] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Indexed: 12/27/2022] Open
Abstract
Progesterone or its analog, one of components of hormone replacement therapy, may attenuate the cardioprotective effects of estrogen. However, the underlying mechanisms have not been fully elucidated. Expression of CD36, a receptor for oxidized LDL (oxLDL) that enhances macrophage/foam cell formation, is activated by the transcription factor peroxisome proliferator-activated receptor γ (PPARγ). CD36 also functions as a fatty acid transporter to influence fatty acid metabolism and the pathophysiological status of several diseases. In this study, we determined that progesterone induced macrophage CD36 expression, which is related to progesterone receptor (PR) activity. Progesterone enhanced cellular oxLDL uptake in a CD36-dependent manner. Mechanistically, progesterone increased PPARγ expression and PPARγ promoter activity in a PR-dependent manner and the binding of PR with the progesterone response element in the PPARγ promoter. Specific deletion of macrophage PPARγ (MφPPARγ KO) expression in mice abolished progesterone-induced macrophage CD36 expression and cellular oxLDL accumulation. We also determined that, associated with gestation and increased serum progesterone levels, CD36 and PPARγ expression in mouse adipose tissue, skeletal muscle, and peritoneal macrophages were substantially activated. Taken together, our study demonstrates that progesterone can play dual pathophysiological roles by activating PPARγ expression, in which progesterone increases macrophage CD36 expression and oxLDL accumulation, a negative effect on atherosclerosis, and enhances the PPARγ-CD36 pathway in adipose tissue and skeletal muscle, a protective effect on pregnancy.
Collapse
Affiliation(s)
| | | | - Yuanli Chen
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China School of Medicine, and
| | - Yan Li
- From the College of Life Sciences
| | - Lei Sun
- From the College of Life Sciences
| | - Ying Liu
- From the College of Life Sciences
| | | | - Miao Yu
- From the College of Life Sciences
| | | | - Jihong Han
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin 300071, China and
| | - Yajun Duan
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin 300071, China and
| |
Collapse
|
62
|
Boghdady A, Arafa UA, Sabet EA, Salama E, El Sharawy A, Elbadry MI. Association between rs1761667 polymorphism of CD36 gene and risk of coronary atherosclerosis in Egyptian population. Cardiovasc Diagn Ther 2016; 6:120-30. [PMID: 27054101 DOI: 10.21037/cdt.2015.12.15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Recent studies have demonstrated that CD36 is involved in the progression of atherosclerosis. Associations between rs1761667 polymorphisms of the CD36 gene and susceptibility to coronary artery disease (CAD) are not obvious. METHODS We studied the relationship between single nucleotide polymorphisms (SNPs), rs1761667 of CD36 gene and the risk of coronary atherosclerosis in a case-control study composed of 71 CAD patients and 76 healthy controls by assessment of allele frequencies and genotype distributions using real-time polymerase chain reaction (PCR) and the allele discrimination technique. Additionally, we detected CD36 expression by flow cytometry. RESULTS The distribution of rs1761667 genotypes between the two groups was significantly different (P<0.001), with the frequency of the AG genotype being significantly higher in the CAD group than in the control group (P<0.001). The expression level of CD36 in the CAD group was significantly higher than that in the control group (P<0.001), with significant differences in the CAD patients with an AG genotype compared with those with an AA and GG genotype (P<0.001). The plasma levels (mg/dL) of low-density lipoprotein (LDL) in the CAD group were much higher than that in the control group (P<0.001). On the other hand, the plasma LDL levels in CAD patients with the AG genotype were remarkably higher than those with the GG and AA genotypes (P=0.046) and AG genotype was significantly more prevalent among type 2 diabetes mellitus (T2DM) and metabolic syndrome (MetS) patients (P<0.05). After adjusted logistic regression analysis, the AG genotype of rs1761667 was associated with an increased risk of CAD (OR=17.97, 95% CI, 3.19-87.85, P=0.001). CONCLUSIONS The AG genotype of the rs1761667 polymorphism in the CD36 gene may be involved in CAD pathogenesis as well as increased body mass index (BMI), T2DM and MetS in the Sohag population of Egypt.
Collapse
Affiliation(s)
- Ahmed Boghdady
- 1 Department of Internal Medicine, Cardiology Division, Cathlab Unit, Sohag University Hospital, Sohag, Egypt ; 2 Department of Internal Medicine, 3 Department of Clinical pathology, 4 Department of Internal Medicine, Hematology Division, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Usama Ahmed Arafa
- 1 Department of Internal Medicine, Cardiology Division, Cathlab Unit, Sohag University Hospital, Sohag, Egypt ; 2 Department of Internal Medicine, 3 Department of Clinical pathology, 4 Department of Internal Medicine, Hematology Division, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Eman A Sabet
- 1 Department of Internal Medicine, Cardiology Division, Cathlab Unit, Sohag University Hospital, Sohag, Egypt ; 2 Department of Internal Medicine, 3 Department of Clinical pathology, 4 Department of Internal Medicine, Hematology Division, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Eman Salama
- 1 Department of Internal Medicine, Cardiology Division, Cathlab Unit, Sohag University Hospital, Sohag, Egypt ; 2 Department of Internal Medicine, 3 Department of Clinical pathology, 4 Department of Internal Medicine, Hematology Division, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Ahmed El Sharawy
- 1 Department of Internal Medicine, Cardiology Division, Cathlab Unit, Sohag University Hospital, Sohag, Egypt ; 2 Department of Internal Medicine, 3 Department of Clinical pathology, 4 Department of Internal Medicine, Hematology Division, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Mahmoud I Elbadry
- 1 Department of Internal Medicine, Cardiology Division, Cathlab Unit, Sohag University Hospital, Sohag, Egypt ; 2 Department of Internal Medicine, 3 Department of Clinical pathology, 4 Department of Internal Medicine, Hematology Division, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| |
Collapse
|
63
|
Alkhatatbeh MJ, Ayoub NM, Mhaidat NM, Saadeh NA, Lincz LF. Soluble cluster of differentiation 36 concentrations are not associated with cardiovascular risk factors in middle-aged subjects. Biomed Rep 2016; 4:642-648. [PMID: 27123261 DOI: 10.3892/br.2016.622] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/01/2016] [Indexed: 11/06/2022] Open
Abstract
Cluster of differentiation 36 (CD36) is involved in the development of atherosclerosis by enhancing macrophage endocytosis of oxidized low-density lipoproteins and foam cell formation. Soluble CD36 (sCD36) was found to be elevated in type 2 diabetic patients and possibly acted as a marker of insulin resistance and atherosclerosis. In young subjects, sCD36 was associated with cardiovascular risk factors including obesity and hypertriglyceridemia. The present study was conducted to further investigate the association between plasma sCD36 and cardiovascular risk factors among middle-aged patients with metabolic syndrome (MetS) and healthy controls. sCD36 concentrations were determined by enzyme-linked immunosorbent assays (ELISA) for 41 patients with MetS and 36 healthy controls. Data for other variables were obtained from patient medical records. sCD36 concentrations were relatively low compared to the majority of other studies and were not significantly different between the MetS group and controls (P=0.17). sCD36 was also not correlated with age, body mass index, glucose, lipid profile, serum electrolytes and blood counts. sCD36 was not significantly different between subjects with obesity, hyperglycemia, dyslipidemia, hypertension or cardiovascular disease, and those without these abnormalities (P>0.05). The inconsistency between results reported in the present study and other studies may be unique to the study population or be a result of the lack of a reliable standardized method for determining absolute sCD36 concentrations. However, further investigations are required to assess CD36 tissue expression in the study population and to assess the accuracy of various commercially available sCD36 ELISA kits. Thus, the availability of a standardized simple sCD36 ELISA that could be performed in any basic laboratory would be more favorable to the specialized flow cytometry methods that detect CD36+ microparticles if it was to be used as a biomarker.
Collapse
Affiliation(s)
- Mohammad J Alkhatatbeh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Nizar M Mhaidat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Nesreen A Saadeh
- Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Lisa F Lincz
- Hunter Haematology Research Group, Calvary Mater Newcastle Hospital, Waratah, NSW 2298, Australia
| |
Collapse
|
64
|
Phang M, Thorne RF, Alkhatatbeh MJ, Garg ML, Lincz LF. Circulating CD36+ microparticles are not altered by docosahexaenoic or eicosapentaenoic acid supplementation. Nutr Metab Cardiovasc Dis 2016; 26:254-260. [PMID: 26803595 DOI: 10.1016/j.numecd.2015.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/20/2015] [Accepted: 12/10/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Circulating microparticles (MP) are the source of a plasma derived form of the scavenger receptor CD36, termed soluble (s)CD36, the levels of which correlate with markers of atherosclerosis and risk of cardiovascular disease. Long chain n-3 polyunsaturated fatty acids have cardioprotective effects that we have previously reported to be gender specific. The aim of this study was to determine if dietary docosahexaenoic acid (DHA) and/or eicosapentaenoic acid (EPA) supplementation affect circulating CD36 + MP levels, and if this occurs differentially in healthy men and women. METHODS AND RESULTS Participants (43M, 51F) aged 39.6 ± 1.7 years received 4 weeks of daily supplementation with DHA rich (200 mg EPA; 1000 mg DHA), EPA rich (1000 mg EPA; 200 mg DHA), or placebo (sunola) oil in a double-blinded, randomised, placebo controlled trial. Plasma CD36 + MP were enumerated by flow cytometry and differences between genders and treatments were evaluated by Student's or paired t-test and one way ANOVA. Males and females had similar levels of CD36 + MP at baseline (mean = 1018 ± 325 vs 980 ± 318; p = 0.577) and these were not significantly changed after DHA (M, p = 0.571; F, p = 0.444) or EPA (M, p = 0.361; F, p = 0.901) supplementation. Likewise, the overall percent change in these levels were not different between supplemented cohorts compared to placebo when all participants were combined (% change in CD36 + MP: DHA = 5.7 ± 37.5, EPA = -3.4 ± 35.4, placebo = -11.5 ± 32.9; p = 0.158) or stratified by gender (M, DHA = -2.6 ± 30.6, EPA = -15.1 ± 20.1, placebo = -21.4 ± 28.7, p = 0.187; F, DHA = 11.7 ± 41.5, EPA = 6.8 ± 42.9, placebo = -2.8 ± 34.7, p = 0.552). CONCLUSION The cardioprotective effects of DHA and EPA do not act through a CD36 + MP mechanism.
Collapse
Affiliation(s)
- M Phang
- Nutraceuticals Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - R F Thorne
- School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW 2258, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - M J Alkhatatbeh
- Clinical Pharmacy Department, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - M L Garg
- Nutraceuticals Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - L F Lincz
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Hunter Haematology Research Group, Calvary Mater Newcastle Hospital, Waratah, NSW 2298, Australia.
| |
Collapse
|
65
|
CD36 is a co-receptor for hepatitis C virus E1 protein attachment. Sci Rep 2016; 6:21808. [PMID: 26898231 PMCID: PMC4761891 DOI: 10.1038/srep21808] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 01/08/2016] [Indexed: 02/06/2023] Open
Abstract
The cluster of differentiation 36 (CD36) is a membrane protein related to lipid metabolism. We show that HCV infection in vitro increased CD36 expression in either surface or soluble form. HCV attachment was facilitated through a direct interaction between CD36 and HCV E1 protein, causing enhanced entry and replication. The HCV co-receptor effect of CD36 was independent of that of SR-BI. CD36 monoclonal antibodies neutralized the effect of CD36 and reduced HCV replication. CD36 inhibitor sulfo-N-succinimidyl oleate (SSO), which directly bound CD36 but not SR-BI, significantly interrupted HCV entry, and therefore inhibited HCV replication. SSO’s antiviral effect was seen only in HCV but not in other viruses. SSO in combination with known anti-HCV drugs showed additional inhibition against HCV. SSO was considerably safe in mice. Conclusively, CD36 interacts with HCV E1 and might be a co-receptor specific for HCV entry; thus, CD36 could be a potential drug target against HCV.
Collapse
|
66
|
Circulating soluble low-density lipoprotein receptor-related protein 1 (sLRP1) concentration is associated with hypercholesterolemia: A new potential biomarker for atherosclerosis. Int J Cardiol 2015; 201:20-9. [DOI: 10.1016/j.ijcard.2015.07.085] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/22/2015] [Accepted: 07/29/2015] [Indexed: 11/22/2022]
|
67
|
Chistiakov DA, Bobryshev YV, Orekhov AN. Macrophage-mediated cholesterol handling in atherosclerosis. J Cell Mol Med 2015; 20:17-28. [PMID: 26493158 PMCID: PMC4717859 DOI: 10.1111/jcmm.12689] [Citation(s) in RCA: 369] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 08/19/2015] [Indexed: 02/06/2023] Open
Abstract
Formation of foam cells is a hallmark at the initial stages of atherosclerosis. Monocytes attracted by pro-inflammatory stimuli attach to the inflamed vascular endothelium and penetrate to the arterial intima where they differentiate to macrophages. Intimal macrophages phagocytize oxidized low-density lipoproteins (oxLDL). Several scavenger receptors (SR), including CD36, SR-A1 and lectin-like oxLDL receptor-1 (LOX-1), mediate oxLDL uptake. In late endosomes/lysosomes of macrophages, oxLDL are catabolysed. Lysosomal acid lipase (LAL) hydrolyses cholesterol esters that are enriched in LDL to free cholesterol and free fatty acids. In the endoplasmic reticulum (ER), acyl coenzyme A: cholesterol acyltransferase-1 (ACAT1) in turn catalyses esterification of cholesterol to store cholesterol esters as lipid droplets in the ER of macrophages. Neutral cholesteryl ester hydrolases nCEH and NCEH1 are involved in a secondary hydrolysis of cholesterol esters to liberate free cholesterol that could be then out-flowed from macrophages by cholesterol ATP-binding cassette (ABC) transporters ABCA1 and ABCG1 and SR-BI. In atherosclerosis, disruption of lipid homoeostasis in macrophages leads to cholesterol accumulation and formation of foam cells.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Division of Laboratory Medicine, Department of Molecular Genetic Diagnostics and Cell Biology, Institute of Pediatrics, Research Center for Children's Health, Moscow, Russia
| | - Yuri V Bobryshev
- Faculty of Medicine and St Vincent's Centre for Applied Medical Research, University of New South Wales, Sydney, NSW, Australia.,School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia.,Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Department of Biophysics, Biological Faculty, Moscow State University, Moscow, Russia
| |
Collapse
|
68
|
Van Dycke KCG, Pennings JLA, van Oostrom CTM, van Kerkhof LWM, van Steeg H, van der Horst GTJ, Rodenburg W. Biomarkers for circadian rhythm disruption independent of time of day. PLoS One 2015; 10:e0127075. [PMID: 25984797 PMCID: PMC4436131 DOI: 10.1371/journal.pone.0127075] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/11/2015] [Indexed: 11/18/2022] Open
Abstract
Frequent shift work causes disruption of the circadian rhythm and might on the long-term result in increased health risk. Current biomarkers evaluating the presence of circadian rhythm disturbance (CRD), including melatonin, cortisol and body temperature, require 24-hr (“around the clock”) measurements, which is tedious. Therefore, these markers are not eligible to be used in large-scale (human) studies. The aim of the present study was to identify universal biomarkers for CRD independent of time of day using a transcriptomics approach. Female FVB mice were exposed to six shifts in a clockwise (CW) and counterclockwise (CCW) CRD protocol and sacrificed at baseline and after 1 shift, 6 shifts, 5 days recovery and 14 days recovery, respectively. At six time-points during the day, livers were collected for mRNA microarray analysis. Using a classification approach, we identified a set of biomarkers able to classify samples into either CRD or non-disrupted based on the hepatic gene expression. Furthermore, we identified differentially expressed genes 14 days after the last shift compared to baseline for both CRD protocols. Non-circadian genes differentially expressed upon both CW and CCW protocol were considered useful, universal markers for CRD. One candidate marker i.e. CD36 was evaluated in serum samples of the CRD animals versus controls. These biomarkers might be useful to measure CRD and can be used later on for monitoring the effectiveness of intervention strategies aiming to prevent or minimize chronic adverse health effects.
Collapse
Affiliation(s)
- Kirsten C. G. Van Dycke
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Genetics, Center for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen L. A. Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Conny T. M. van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Linda W. M. van Kerkhof
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Harry van Steeg
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Wendy Rodenburg
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- * E-mail:
| |
Collapse
|
69
|
Krzystolik A, Dziedziejko V, Safranow K, Kurzawski G, Rać M, Sagasz-Tysiewicz D, Poncyljusz W, Jakubowska K, Chlubek D, Rać ME. Is plasma soluble CD36 associated with cardiovascular risk factors in early onset coronary artery disease patients? Scandinavian Journal of Clinical and Laboratory Investigation 2015; 75:398-406. [DOI: 10.3109/00365513.2015.1031693] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Grzegorz Kurzawski
- Department of Genetics and Pathomorphology, Pomeranian Medical University, Szczecin, Poland
| | - Michał Rać
- Department of Diagnostic Imaging and Interventional Radiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Wojciech Poncyljusz
- Department of Interventional Radiology, Pomeranian Medical University, Szczecin, Poland
| | - Katarzyna Jakubowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Monika E. Rać
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
70
|
Hjuler Nielsen M, Irvine H, Vedel S, Raungaard B, Beck-Nielsen H, Handberg A. Elevated atherosclerosis-related gene expression, monocyte activation and microparticle-release are related to increased lipoprotein-associated oxidative stress in familial hypercholesterolemia. PLoS One 2015; 10:e0121516. [PMID: 25875611 PMCID: PMC4395270 DOI: 10.1371/journal.pone.0121516] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/02/2015] [Indexed: 02/06/2023] Open
Abstract
Objective Animal and in vitro studies have suggested that hypercholesterolemia and increased oxidative stress predisposes to monocyte activation and enhanced accumulation of oxidized LDL cholesterol (oxLDL-C) through a CD36-dependent mechanism. The aim of this study was to investigate the hypothesis that elevated oxLDL-C induce proinflammatory monocytes and increased release of monocyte-derived microparticles (MMPs), as well as up-regulation of CD36, chemokine receptors and proinflammatory factors through CD36-dependent pathways and that this is associated with accelerated atherosclerosis in subjects with heterozygous familial hypercholesterolemia (FH), in particular in the presence of Achilles tendon xanthomas (ATX). Approach and Results We studied thirty FH subjects with and without ATX and twenty-three healthy control subjects. Intima-media thickness (IMT) and Achilles tendon (AT) thickness were measured by ultrasonography. Monocyte classification and MMP analysis were performed by flow cytometry. Monocyte expression of genes involved in atherosclerosis was determined by quantitative PCR. IMT and oxLDL-C were increased in FH subjects, especially in the presence of ATX. In addition, FH subjects had elevated proportions of intermediate CD14++CD16+ monocytes and higher circulating MMP levels. Stepwise linear regression identified oxLDL-C, gender and intermediate monocytes as predictors of MMPs. Monocyte expression of pro-atherogenic and pro-inflammatory genes regulated by oxLDL-C-CD36 interaction was increased in FH, especially in ATX+ subjects. Monocyte chemokine receptor CX3CR1 was identified as an independent contributor to IMT. Conclusions Our data support that lipoprotein-associated oxidative stress is involved in accelerated atherosclerosis in FH, particularly in the presence of ATX, by inducing pro-inflammatory monocytes and increased release of MMPs along with elevated monocyte expression of oxLDL-C-induced atherosclerosis-related genes.
Collapse
Affiliation(s)
- Morten Hjuler Nielsen
- Danish PhD School of Molecular Metabolism, University of Southern Denmark, Odense, Denmark
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- * E-mail:
| | - Helle Irvine
- Department of Medicine and Cardiology A, Aarhus University Hospital, Aarhus, Denmark
| | - Simon Vedel
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | - Bent Raungaard
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
71
|
Shiju TM, Mohan V, Balasubramanyam M, Viswanathan P. Soluble CD36 in plasma and urine: a plausible prognostic marker for diabetic nephropathy. J Diabetes Complications 2015; 29:400-406. [PMID: 25619588 DOI: 10.1016/j.jdiacomp.2014.12.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 02/05/2023]
Abstract
AIMS This study was designed to analyze the level of soluble CD36 (sCD36) in both plasma and urine of type 2 diabetic patients with and without microalbuminuria/macroalbuminuria. METHODS Study subjects (n=20 each) comprised of those with normal glucose tolerance, type 2 diabetes (T2DM) with normoalbuminiria, T2DM with microalbuminuria and T2DM with macroalbuminuria. The biochemical parameters were analyzed using auto-analyzer, and the level of sCD36 was estimated using an in-house Sandwich ELISA. RESULTS The presence of sCD36 has been identified for the first time in the urine sample. Significant increase in the level of sCD36 was observed in both plasma and urine of diabetic patients with microalbuminuria (P<0.01) and macroalbuminuria (P<0.001). Positive correlation of sCD36 with the kidney markers such as urea, creatinine and eGFR confirmed the association of sCD36 with kidney damage in diabetic patients. Microalbuminuria, which is clinically used as a biomarker for nephropathy showed a strong positive correlation with urine sCD36 (r=0.642; P<0.001) and plasma sCD36 (r=0.498; P<0.001) in Pearson correlation analysis, which was further substantiated in stepwise multiple regression analysis. CONCLUSIONS Our study implies a plausible prognostic/adjuvant biomarker role of soluble CD36 for diabetic nephropathy.
Collapse
Affiliation(s)
- Thomas Michael Shiju
- Renal Research Lab, Centre for Bio medical Research, School of Bio Sciences and Technology, VIT University, Vellore 632 014, India
| | - Viswanathan Mohan
- Department of Cell and Molecular Biology, Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Gopalapuram, Chennai, India
| | - Muthuswamy Balasubramanyam
- Department of Cell and Molecular Biology, Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Gopalapuram, Chennai, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Bio medical Research, School of Bio Sciences and Technology, VIT University, Vellore 632 014, India.
| |
Collapse
|
72
|
Kasak L, Rull K, Vaas P, Teesalu P, Laan M. Extensive load of somatic CNVs in the human placenta. Sci Rep 2015; 5:8342. [PMID: 25666259 PMCID: PMC4914949 DOI: 10.1038/srep08342] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/15/2015] [Indexed: 11/09/2022] Open
Abstract
Placenta is a temporary, but indispensable organ in mammalian pregnancy. From its basic nature, it exhibits highly invasive tumour-like properties facilitating effective implantation through trophoblast cell proliferation and migration, and a critical role in pregnancy success. We hypothesized that similarly to cancer, somatic genomic rearrangements are promoted in the support of placental function. Here we present the first profiling of copy number variations (CNVs) in human placental genomes, showing an extensive load of somatic CNVs, especially duplications and suggesting that this phenomenon may be critical for normal gestation. Placental somatic CNVs were significantly enriched in genes involved in cell adhesion, immunity, embryonic development and cell cycle. Overrepresentation of imprinted genes in somatic duplications suggests that amplified gene copies may represent an alternative mechanism to support parent-of-origin specific gene expression. Placentas from pregnancy complications exhibited significantly altered CNV profile compared to normal gestations, indicative to the clinical implications of the study.
Collapse
Affiliation(s)
- Laura Kasak
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Riia St. 23, Tartu 51010, Estonia
| | - Kristiina Rull
- 1] Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Riia St. 23, Tartu 51010, Estonia [2] Department of Obstetrics and Gynaecology, University of Tartu, Puusepa St. 8, Tartu 51014, Estonia [3] Women's Clinic of Tartu University Hospital, Puusepa St. 8, Tartu 51014, Estonia
| | - Pille Vaas
- 1] Department of Obstetrics and Gynaecology, University of Tartu, Puusepa St. 8, Tartu 51014, Estonia [2] Women's Clinic of Tartu University Hospital, Puusepa St. 8, Tartu 51014, Estonia
| | - Pille Teesalu
- 1] Department of Obstetrics and Gynaecology, University of Tartu, Puusepa St. 8, Tartu 51014, Estonia [2] Women's Clinic of Tartu University Hospital, Puusepa St. 8, Tartu 51014, Estonia
| | - Maris Laan
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Riia St. 23, Tartu 51010, Estonia
| |
Collapse
|
73
|
Ellis J, Lange EM, Li J, Dupuis J, Baumert J, Walston JD, Keating BJ, Durda P, Fox ER, Palmer CD, Meng YA, Young T, Farlow DN, Schnabel RB, Marzi CS, Larkin E, Martin LW, Bis JC, Auer P, Ramachandran VS, Gabriel SB, Willis MS, Pankow JS, Papanicolaou GJ, Rotter JI, Ballantyne CM, Gross MD, Lettre G, Wilson JG, Peters U, Koenig W, Tracy RP, Redline S, Reiner AP, Benjamin EJ, Lange LA. Large multiethnic Candidate Gene Study for C-reactive protein levels: identification of a novel association at CD36 in African Americans. Hum Genet 2014; 133:985-95. [PMID: 24643644 PMCID: PMC4104766 DOI: 10.1007/s00439-014-1439-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/06/2014] [Indexed: 10/25/2022]
Abstract
C-reactive protein (CRP) is a heritable biomarker of systemic inflammation and a predictor of cardiovascular disease (CVD). Large-scale genetic association studies for CRP have largely focused on individuals of European descent. We sought to uncover novel genetic variants for CRP in a multiethnic sample using the ITMAT Broad-CARe (IBC) array, a custom 50,000 SNP gene-centric array having dense coverage of over 2,000 candidate CVD genes. We performed analyses on 7,570 African Americans (AA) from the Candidate gene Association Resource (CARe) study and race-combined meta-analyses that included 29,939 additional individuals of European descent from CARe, the Women's Health Initiative (WHI) and KORA studies. We observed array-wide significance (p < 2.2 × 10(-6)) for four loci in AA, three of which have been reported previously in individuals of European descent (IL6R, p = 2.0 × 10(-6); CRP, p = 4.2 × 10(-71); APOE, p = 1.6 × 10(-6)). The fourth significant locus, CD36 (p = 1.6 × 10(-6)), was observed at a functional variant (rs3211938) that is extremely rare in individuals of European descent. We replicated the CD36 finding (p = 1.8 × 10(-5)) in an independent sample of 8,041 AA women from WHI; a meta-analysis combining the CARe and WHI AA results at rs3211938 reached genome-wide significance (p = 1.5 × 10(-10)). In the race-combined meta-analyses, 13 loci reached significance, including ten (CRP, TOMM40/APOE/APOC1, HNF1A, LEPR, GCKR, IL6R, IL1RN, NLRP3, HNF4A and BAZ1B/BCL7B) previously associated with CRP, and one (ARNTL) previously reported to be nominally associated with CRP. Two novel loci were also detected (RPS6KB1, p = 2.0 × 10(-6); CD36, p = 1.4 × 10(-6)). These results highlight both shared and unique genetic risk factors for CRP in AA compared to populations of European descent.
Collapse
Affiliation(s)
- Jaclyn Ellis
- Department of Genetics, University of North Carolina, 5112 Genetic Medicine Bldg., Chapel Hill, NC, 27599-7264, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Park YM. CD36, a scavenger receptor implicated in atherosclerosis. Exp Mol Med 2014; 46:e99. [PMID: 24903227 PMCID: PMC4081553 DOI: 10.1038/emm.2014.38] [Citation(s) in RCA: 377] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 03/17/2014] [Accepted: 04/08/2014] [Indexed: 12/17/2022] Open
Abstract
CD36 is a membrane glycoprotein that is present on various types of cells, including monocytes, macrophages, microvascular endothelial cells, adipocytes and platelets. Macrophage CD36 participates in atherosclerotic arterial lesion formation through its interaction with oxidized low-density lipoprotein (oxLDL), which triggers signaling cascades for inflammatory responses. CD36 functions in oxLDL uptake and foam cell formation, which is the initial critical stage of atherosclerosis. In addition, oxLDL via CD36 inhibits macrophage migration, which may be a macrophage-trapping mechanism in atherosclerotic lesions. The role of CD36 was examined in in vitro studies and in vivo experiments, which investigated various functions of CD36 in atherosclerosis and revealed that CD36 deficiency reduces atherosclerotic lesion formation. Platelet CD36 also promotes atherosclerotic inflammatory processes and is involved in thrombus formation after atherosclerotic plaque rupture. Because CD36 is an essential component of atherosclerosis, defining the function of CD36 and its corresponding signaling pathway may lead to a new treatment strategy for atherosclerosis.
Collapse
Affiliation(s)
- Young Mi Park
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
- Ewha Global Top 5 Research Program, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
75
|
Kim E, Tolhurst AT, Cho S. Deregulation of inflammatory response in the diabetic condition is associated with increased ischemic brain injury. J Neuroinflammation 2014; 11:83. [PMID: 24886035 PMCID: PMC4017808 DOI: 10.1186/1742-2094-11-83] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/26/2014] [Indexed: 01/04/2023] Open
Abstract
Background Although elicited inflammation contributes to tissue injury, a certain level of inflammation is necessary for subsequent tissue repair/remodeling. Diabetes, a chronic low-grade inflammatory state, is a predisposing risk factor for stroke. The condition is associated with delayed wound healing, presumably due to disrupted inflammatory responses. With inclusion of the diabetic condition in an experimental animal model of stroke, this study investigates whether the condition alters inflammatory response and influences stroke-induced brain injury. Methods C57BL/6 mice were fed a diabetic diet (DD) for 8 weeks to induce an experimental diabetic condition or a normal diet (ND) for the same duration. Gene expression of inflammatory factors including monocyte chemoattractant protein-1 (MCP-1), interleukin-6 (IL-6), CCR2, and CD36 was assessed in the peripheral immune cells and brains of normal and diabetic mice before and after focal cerebral ischemia. The expression of these factors was also determined in lipopolysaccharide (LPS)-treated cultured normal and diabetic macrophages. Ischemic outcome was assessed in these mice at 3 days post-ischemia. Results DD intervention in mice resulted in obesity and elevated insulin and glucose level in the blood. The peritoneal immune cells from the diabetic mice showed higher MCP-1 mRNA levels before and after stroke. Compared to normal mice, diabetic mice showed reduced MCP-1, IL-6, and CCR2 gene expression in the brain at 6 h post-ischemia. LPS-stimulated inflammatory responses were also reduced in the diabetic macrophages. The diabetic mice showed larger infarct size and percent swelling. Conclusions These results showed that diabetic conditions deregulate acute inflammatory response and that the condition is associated with increased stroke-induced injury. The study suggests that interventions aimed at restoring appropriate inflammatory response in peripheral immune cells/macrophages may be beneficial in reducing stroke-induced brain injury in subjects with chronic inflammatory conditions.
Collapse
Affiliation(s)
| | | | - Sunghee Cho
- Burke-Cornell Medical Research Institute, White Plains, NY 10605, USA.
| |
Collapse
|
76
|
Ramos-Arellano LE, Muñoz-Valle JF, De la Cruz-Mosso U, Salgado-Bernabé AB, Castro-Alarcón N, Parra-Rojas I. Circulating CD36 and oxLDL levels are associated with cardiovascular risk factors in young subjects. BMC Cardiovasc Disord 2014; 14:54. [PMID: 24766787 PMCID: PMC4012526 DOI: 10.1186/1471-2261-14-54] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 04/23/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) results from a combination of abnormalities in lipoprotein metabolism, oxidative stress, chronic inflammation, and susceptibility to thrombosis. Atherosclerosis is the major cause of CVD. CD36 has been shown to play a critical role in the development of atherosclerotic lesions by its capacity to bind and promote endocytosis of oxidized low-density lipoprotein (oxLDL) and is implicated in the formation of foam cells. The purpose of this research was to evaluate whether there is an association of sCD36 and oxLDL levels with cardiovascular risk factors in young subjects. METHODS A total of 188 subjects, 18 to 25 years old, 133 normal-weight and 55 obese subjects from the state of Guerrero, Mexico were recruited in the study. The lipid profile and glucose levels were measured by enzymatic colorimetric assays. Enzyme-linked immunosorbant assays (ELISA) for oxLDL and sCD36 were performed. Statistical analyses of data were performed with Wilcoxon- Mann Whitney and chi-square tests as well as with multinomial regression. RESULTS TC, LDL-C, TG, oxLDL and sCD36 levels were higher in obese subjects than in normal-weight controls, as well as, monocyte and platelet counts (P < 0.05). Obese subjects had 5.8 times higher risk of sCD36 in the third tertil (>97.8 ng/mL) than normal-weight controls (P = 0.014), and 7.4 times higher risk of oxLDL levels in third tertile (>48 U/L) than control group. The subjects with hypercholesterolemia, hypertriglyceridemia, fasting impaired LDL-C had a higher risk of oxLDL levels in the third tertile (>48 U/L) than the control group (P < 0.05). CONCLUSIONS Circulating CD36 and oxLDL levels are associated with cardiovascular risk factors in young subjects and may be potential early markers for cardiovascular disease (CVD).
Collapse
Affiliation(s)
| | | | | | | | | | - Isela Parra-Rojas
- Laboratorio de Investigación en Obesidad y Diabetes, Unidad Académica de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México.
| |
Collapse
|
77
|
Knøsgaard L, Thomsen SB, Støckel M, Vestergaard H, Handberg A. Circulating sCD36 is associated with unhealthy fat distribution and elevated circulating triglycerides in morbidly obese individuals. Nutr Diabetes 2014; 4:e114. [PMID: 24710072 PMCID: PMC4007154 DOI: 10.1038/nutd.2014.11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 02/20/2014] [Indexed: 12/13/2022] Open
Abstract
Background: The recently identified circulating sCD36 has been proposed to reflect tissue CD36 expression, and is upregulated in case of obesity, insulin resistance and hepatic steatosis. The aim of this study was to explore the effect of weight loss secondary to bariatric surgery in relation to sCD36 among morbidly obese individuals. Furthermore, we investigated the levels of sCD36 in relation to obesity-related metabolic complications, low-grade inflammation and fat distribution. Methods: Twenty morbidly obese individuals (body mass index (BMI) 43.0±5.4 kg m−2) with a referral to Roux-en-Y gastric bypass were included. Anthropometric measurements and fasting blood samples were collected at a preoperative baseline visit and 3 months after surgery. sCD36 was measured by an in-house assay, whereas insulin sensitivity and the hepatic fat accumulation were estimated by the homeostasis model assessment (HOMA-%S) and liver fat percentage (LF%), respectively. Results: Postoperatively, BMI was reduced by 20% to 34.3±5.2 kg m−2 (P<0.001). sCD36 was reduced by 31% (P=0.001) and improvements were observed in the amount of fat mass (P<0.001), truncal fat mass (P<0.001), circulating triglycerides (P=0.001), HOMA-%S (P=0.007), LF% (P=0.001) and the inflammatory marker high-sensitive C-reactive protein (P=0.005). sCD36 correlated with triglycerides (ρ=0.523, P=0.001) and truncal fat mass (ρ=0.357, P=0.026), and triglycerides were found to be an independent predictor of sCD36. At baseline, participants with the metabolic syndrome had a higher LF% and higher levels of the inflammatory biomarker YKL-40 (P=0.003 and P=0.014) as well as a tendency towards higher levels of sCD36. Conclusion: sCD36 was reduced by weight loss and associated with an unhealthy fat accumulation and circulating triglycerides, which support the proposed role of sCD36 as a biochemical marker of obesity-related metabolic complications and risks.
Collapse
Affiliation(s)
- L Knøsgaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - S B Thomsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M Støckel
- Department of Surgery, Copenhagen University Hospital, Herlev, Denmark
| | - H Vestergaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - A Handberg
- 1] Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark [2] Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
78
|
García-Monzón C, Lo Iacono O, Crespo J, Romero-Gómez M, García-Samaniego J, Fernández-Bermejo M, Domínguez-Díez A, Rodríguez de Cía J, Sáez A, Porrero JL, Vargas-Castrillón J, Chávez-Jiménez E, Soto-Fernández S, Díaz A, Gallego-Durán R, Madejón A, Miquilena-Colina ME. Increased soluble CD36 is linked to advanced steatosis in nonalcoholic fatty liver disease. Eur J Clin Invest 2014; 44:65-73. [PMID: 24134687 DOI: 10.1111/eci.12192] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 10/15/2013] [Indexed: 12/24/2022]
Abstract
BACKGROUND Soluble CD36 (sCD36) clusters with insulin resistance, but no evidence exists on its relationship with hepatic fat content. We determined sCD36 to assess its link to steatosis in nonalcoholic fatty liver disease (NAFLD) and chronic hepatitis C (CHC) patients. MATERIALS AND METHODS Two hundred and twenty-seven NAFLD, eighty-seven CHC, and eighty-five patients with histologically normal liver (NL) were studied. Steatosis was graded by Kleiner's histological scoring system. Serum sCD36 and hepatic CD36 expression was assessed by immunoassay and immunohistochemistry, respectively. RESULTS In NAFLD, serum sCD36 levels were significantly higher in simple steatosis than in NL (361.4 ± 286.4 vs. 173.9 ± 137.4 pg/mL, respectively; P < 0.001), but not in steatohepatitis (229.6 ± 202.5 pg/mL; P = 0.153). In CHC, serum sCD36 levels were similar regardless of the absence (428.7 ± 260.3 pg/mL) or presence of steatosis (387.2 ± 283.6 pg/mL; P = 0.173). A progressive increase in serum sCD36 values was found in NAFLD depending on the histological grade of steatosis (P < 0.001), but not in CHC (P = 0.151). Serum sCD36 concentrations were independently associated with advanced steatosis in NAFLD when adjusted by demographic and anthropometric features [odds ratio (OR), 1.001; 95% confidence interval (CI), 1.000 to 1.002; P = 0.021] and by metabolic variables (OR, 1.002; 95% CI, 1.000 to 1.003; P = 0.001). Interestingly, a significant correlation was observed between hepatic CD36 and serum sCD36 (ρ = 0.499, P < 0.001). CONCLUSIONS Increased serum sCD36 is an independent factor associated with advanced steatosis in NAFLD.
Collapse
Affiliation(s)
- Carmelo García-Monzón
- Liver Research Unit, University Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa, CIBEREHD, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Wilhelmsen P, Kjær J, Thomsen KL, Nielsen CT, Dige A, Maniecki MB, Heegaard N, Grønbæk H, Dahlerup J, Handberg A. Elevated platelet expression of CD36 may contribute to increased risk of thrombo-embolism in active inflammatory bowel disease. Arch Physiol Biochem 2013; 119:202-8. [PMID: 23862574 DOI: 10.3109/13813455.2013.808671] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CONTEXT Inflammatory bowel disease (IBD) induces increased risk of thrombo-embolism. CD36 is involved in platelet activation, glucose metabolism and inflammation. OBJECTIVE The relationship between CD36 expression on platelets and monocytes, plasma sCD36, and CD36-positive platelet-derived microparticles (PDMPs) and inflammation in both active IBD and after one week of anti-tumour necrosis alpha antibody (anti-TNF) treatment was investigated. MATERIAL AND METHODS Patients with exacerbation of Crohn's disease (n = 8) or ulcerative colitis (n = 5) and 13 healthy controls were enrolled. Seven patients underwent anti-TNF treatment for one week. Platelet, monocyte, and PDMP-CD36 were measured by flow-cytometry. RESULTS Platelet CD36 expression was 34% higher in patients, and correlated with insulin resistance and fasting glucose. sCD36 was 37% lower and restored after anti-TNF treatment. CONCLUSION Elevated platelet CD36 expression may contribute to increased risk of thrombo-embolism in active IBD. This may not entirely be attributed to inflammation and secondary insulin resistance may play a role.
Collapse
Affiliation(s)
- Peter Wilhelmsen
- Department of Clinical Biochemistry, Aarhus University Hospital , Aarhus , Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Bacon S, Kyithar MP, Schmid J, Costa Pozza A, Handberg A, Byrne MM. Circulating CD36 is reduced in HNF1A-MODY carriers. PLoS One 2013; 8:e74577. [PMID: 24069322 PMCID: PMC3771933 DOI: 10.1371/journal.pone.0074577] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 08/05/2013] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Premature atherosclerosis is a significant cause of morbidity and mortality in type 2 diabetes mellitus. Maturity onset diabetes of the young (MODY) accounts for approximately 2% of all diabetes, with mutations in the transcription factor; hepatocyte nuclear factor 1 alpha (HNF1A) accounting for the majority of MODY cases. There is somewhat limited data available on the prevalence of macrovascular disease in HNF1A-MODY carriers with diabetes. Marked insulin resistance and the associated dyslipidaemia are not clinical features of HNF1A-MODY carriers. The scavenger protein CD36 has been shown to play a substantial role in the pathogenesis of atherosclerosis, largely through its interaction with oxidised LDL. Higher levels of monocyte CD36 and plasma CD36(sCD36) are seen to cluster with insulin resistance and diabetes. The aim of this study was to determine levels of sCD36 in participants with HNF1A-MODY diabetes and to compare them with unaffected normoglycaemic family members and participants with type 2 diabetes mellitus. METHODS We recruited 37 participants with HNF1A-MODY diabetes and compared levels of sCD36 with BMI-matched participants with type 2 diabetes mellitus and normoglycaemic HNF1A-MODY negative family controls. Levels of sCD36 were correlated with phenotypic and biochemical parameters. RESULTS HNF1A-MODY participants were lean, normotensive, with higher HDL and lower triglyceride levels when compared to controls and participants with type 2 diabetes mellitus. sCD36 was also significantly lower in HNF1A-MODY participants when compared to both the normoglycaemic family controls and to lean participants with type 2 diabetes mellitus. CONCLUSION In conclusion, sCD36 is significantly lower in lean participants with HNF1A-MODY diabetes when compared to weight-matched normoglycaemic familial HNF1A-MODY negative controls and to lean participants with type 2 diabetes mellitus. Lower levels of this pro-atherogenic marker may result from the higher HDL component in the lipid profile of HNF1A-MODY participants.
Collapse
Affiliation(s)
- Siobhan Bacon
- Department of Endocrinology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Ma P. Kyithar
- Department of Endocrinology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Jasmin Schmid
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Andre Costa Pozza
- Department of Endocrinology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Maria M. Byrne
- Department of Endocrinology, Mater Misericordiae University Hospital, Dublin, Ireland
- * E-mail:
| |
Collapse
|
81
|
Himoto T, Tani J, Miyoshi H, Morishita A, Yoneyama H, Kurokohchi K, Inukai M, Masugata H, Goda F, Senda S, Haba R, Ueno M, Yamaoka G, Masaki T. Investigation of the factors associated with circulating soluble CD36 levels in patients with HCV-related chronic liver disease. Diabetol Metab Syndr 2013; 5:51. [PMID: 24016701 PMCID: PMC3846866 DOI: 10.1186/1758-5996-5-51] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/02/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND CD36, a class B scavenger receptor, participates in the pathogenesis of metabolic dysregulation such as insulin resistance, hepatic steatosis, and atherosclerosis. Persistent hepatitis C virus (HCV) infection often evokes these metabolic abnormalities. The primary purpose of this study was to investigate the role of CD36 in the pathogenesis of insulin resistance and hepatic steatosis caused by chronic HCV infection. METHODS Forty-five patients with HCV-related chronic liver disease (CLD-C) were enrolled in this study. CD36 expression in the liver specimen was examined by an immunohistochemical procedure. The concentrations of circulating soluble form of CD36 (sCD36) and oxLDL were determined by the enzyme-linked innunosorbent assay. Insulin resistance was estimated by the values of HOMA-IR. RESULTS Moderate to extensive hepatic CD36 expression was observed in the sinusoids of all enrolled CLD-C patients. CD36-positive sinusoids appeared to be identical to Kupffer cells. The severity of CD36 expression in the hepatic sinusoids was significantly correlated with the sCD36 level in sera of patients with CLD-C. The serum sCD36 levels were significantly correlated with body mass index and serum oxLDL levels in those patients. However, the serum sCD36 concentrations were independent of the values of HOMA-IR and the severity of hepatic steatosis. CONCLUSIONS These data suggest that the serum sCD36 levels reflect the severity of CD36 expression on the Kupffer cells in patients with CLD-C, and that the serum sCD36 levels were associated with obesity, although the levels were independent of insulin resistance and hepatic steatosis in those patients.
Collapse
Affiliation(s)
- Takashi Himoto
- Department of Integrated Medicine, Kagawa University School of Medicine, 1750-1, Ikenobe, Miki-Cho, Kagawa 7610-79, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Kagawa University School of Medicine, Kagawa, Japan
| | - Hisaaki Miyoshi
- Department of Gastroenterology and Neurology, Kagawa University School of Medicine, Kagawa, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University School of Medicine, Kagawa, Japan
| | - Hirohito Yoneyama
- Department of Gastroenterology and Neurology, Kagawa University School of Medicine, Kagawa, Japan
| | - Kazutaka Kurokohchi
- Department of Gastroenterology and Neurology, Kagawa University School of Medicine, Kagawa, Japan
| | - Michio Inukai
- Department of Integrated Medicine, Kagawa University School of Medicine, 1750-1, Ikenobe, Miki-Cho, Kagawa 7610-79, Japan
| | - Hisashi Masugata
- Department of Integrated Medicine, Kagawa University School of Medicine, 1750-1, Ikenobe, Miki-Cho, Kagawa 7610-79, Japan
| | - Fuminori Goda
- Department of Integrated Medicine, Kagawa University School of Medicine, 1750-1, Ikenobe, Miki-Cho, Kagawa 7610-79, Japan
| | - Shoichi Senda
- Department of Integrated Medicine, Kagawa University School of Medicine, 1750-1, Ikenobe, Miki-Cho, Kagawa 7610-79, Japan
| | - Reiji Haba
- Department of Diagnosis Pathology, Kagawa University School of Medicine, Kagawa, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Kagawa University School of Medicine, Kagawa, Japan
| | - Genji Yamaoka
- Department of Clinical Laboratory, Hospital of Kagawa University School of Medicine, Kagawa, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University School of Medicine, Kagawa, Japan
| |
Collapse
|
82
|
Venø SK, Nielsen MRS, Lundbye-Christensen S, Schmidt EB, Handberg A. The effect of low-dose marine n-3 fatty acids on plasma levels of sCD36 in overweight subjects: a randomized, double-blind, placebo-controlled trial. Mar Drugs 2013; 11:3324-34. [PMID: 23999661 PMCID: PMC3806470 DOI: 10.3390/md11093324] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 08/01/2013] [Accepted: 08/08/2013] [Indexed: 12/31/2022] Open
Abstract
CD36 is a scavenger receptor involved in lipid uptake and inflammation. Recently, non-cell-bound CD36 (sCD36) was identified in plasma and suggested to be a marker of lipid accumulation in the vessel wall. Marine n-3 polyunsaturated fatty acids (PUFA) may have cardioprotective effects. This study evaluated the effect of marine n-3 PUFA on sCD36 levels in overweight subjects. Fifty overweight subjects were randomized to 1.1 g of n-3 PUFA or 2 g of olive oil daily for six weeks. Neutrophils were isolated at baseline and after six weeks of treatment while an adipose tissue biopsy was obtained at baseline. The content of n-3 PUFA in adipose tissue and neutrophils was analyzed by gas chromatography, while plasma levels of sCD36 were determined using an enzyme-linked immunosorbent assay (ELISA). After six weeks of supplement plasma sCD36 did not differ between supplements (P = 0.18). There was no significant correlation between plasma sCD36 levels and n-3 PUFA in neutrophils at baseline (r = −0.02, P = 0.88), after six weeks supplement (r = −0.03, P = 0.85) or in adipose tissue (r = 0.14, P = 0.34). This study therefore does not provide evidence for a cardioprotective effect of n-3 PUFA acting through a CD36-dependent mechanism.
Collapse
Affiliation(s)
- Stine Krogh Venø
- Department of Cardiology, Center for Cardiovascular Research, Aalborg University Hospital, Aalborg 9000, Denmark; E-Mails: (M.R.S.N.); (S.L.-C.); (E.B.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +45-993-268-99; Fax: +45-993-268-13
| | - Michael René Skjelbo Nielsen
- Department of Cardiology, Center for Cardiovascular Research, Aalborg University Hospital, Aalborg 9000, Denmark; E-Mails: (M.R.S.N.); (S.L.-C.); (E.B.S.)
| | - Søren Lundbye-Christensen
- Department of Cardiology, Center for Cardiovascular Research, Aalborg University Hospital, Aalborg 9000, Denmark; E-Mails: (M.R.S.N.); (S.L.-C.); (E.B.S.)
| | - Erik Berg Schmidt
- Department of Cardiology, Center for Cardiovascular Research, Aalborg University Hospital, Aalborg 9000, Denmark; E-Mails: (M.R.S.N.); (S.L.-C.); (E.B.S.)
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg 9000, Denmark; E-Mail:
| |
Collapse
|
83
|
Petta S, Handberg A, Marchesini G, Cammà C, Di Marco V, Cabibi D, Macaluso FS, Craxì A. High sCD36 plasma level is associated with steatosis and its severity in patients with genotype 1 chronic hepatitis C. J Viral Hepat 2013; 20:174-82. [PMID: 23383656 DOI: 10.1111/j.1365-2893.2012.01641.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Soluble CD36 (sCD36) plasma levels, a known marker of cardiometabolic disorders, are associated with surrogate markers of steatosis, while experimental and human studies show a link between CD36 expression in the liver and steatosis. In a cohort of patients with genotype 1 chronic hepatitis C (G1 CHC), we tested the association of sCD36 plasma levels with host and viral factors and sustained virological response (SVR). One hundred and seventy-five consecutive biopsy-proven patients were studied. sCD36 plasma levels were assessed by an in-house ELISA. All biopsies were scored by one pathologist for staging and grading (Scheuer) and graded for steatosis, which was considered moderate-severe if ≥20%. Patients underwent standard of care therapy with pegylated interferon and ribavirin. The severity of steatosis progressively increased according to sCD36 quartiles (P = 0.02); total and low-density lipoprotein (LDL) cholesterol levels were significantly higher in patients in the lower quartile compared to all the others. Gamma-glutamyl transferase (P = 0.02), homoeostasis model assessment (HOMA) score (P = 0.002) and sCD36 (P = 0.04) were independently associated with the severity of steatosis as continuous variable. Multivariate logistic regression analysis showed that HOMA (OR 1.243, 95% CI 1.04-1.484, P = 0.01) and sCD36 (OR 1.445, 95%CI 1.135-1.839, P = 0.003) were independently linked to steatosis ≥20%. No association was found between sCD36 and SVR. CD36 is linked to steatosis and insulin resistance in patients with G1 CHC, but does not predict response to treatment. The potential of sCD36 as a surrogate marker of steatosis should be further investigated.
Collapse
Affiliation(s)
- S Petta
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Palermo, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Abstract
Objective: Elevated plasma levels of the fatty acid transporter, CD36, have been shown to constitute a novel biomarker for type 2 diabetes mellitus (T2DM). We recently reported such circulating CD36 to be entirely associated with cellular microparticles (MPs) and aim here to determine the absolute levels and cellular origin(s) of these CD36+MPs in persons with T2DM. Design: An ex vivo case-control study was conducted using plasma samples from 33 obese individuals with T2DM (body mass index (BMI)=39.9±6.4 kg m−2; age=57±9 years; 18 male:15 female) and age- and gender-matched lean and obese non-T2DM controls (BMI=23.6±1.8 kg m−2 and 33.5±5.9 kg m−2, respectively). Flow cytometry was used to analyse surface expression of CD36 together with tissue-specific markers: CD41, CD235a, CD14, CD105 and phosphatidyl serine on plasma MPs. An enzyme-linked immunosorbent assay was used to quantify absolute CD36 protein concentrations. Results: CD36+MP levels were significantly higher in obese people with T2DM (P<0.00001) and were primarily derived from erythrocytes (CD235a+=35.8±14.6%); although this did not correlate with haemoglobin A1c. By contrast, the main source of CD36+MPs in non-T2DM individuals was endothelial cells (CD105+=40.9±8.3% and 33.9±8.3% for lean and obese controls, respectively). Across the entire cohort, plasma CD36 protein concentration varied from undetectable to 22.9 μg ml−1 and was positively correlated with CD36+MPs measured by flow cytometry (P=0.0006) but only weakly associated with the distribution of controls and T2DM (P=0.021). Multivariate analysis confirmed that plasma CD36+MP levels were a much better biomarker for diabetes than CD36 protein concentration (P=0.009 vs P=0.398, respectively). Conclusions: Both the levels and cellular profile of CD36+MPs differ in T2DM compared with controls, suggesting that these specific vesicles could represent distinct biological vectors contributing to the pathology of the disease.
Collapse
|
85
|
Syed Ikmal SIQ, Zaman Huri H, Vethakkan SR, Wan Ahmad WA. Potential biomarkers of insulin resistance and atherosclerosis in type 2 diabetes mellitus patients with coronary artery disease. Int J Endocrinol 2013; 2013:698567. [PMID: 24282409 PMCID: PMC3824310 DOI: 10.1155/2013/698567] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 09/10/2013] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes mellitus patients with coronary artery disease have become a major public health concern. The occurrence of insulin resistance accompanied with endothelial dysfunction worsens the state of atherosclerosis in type 2 diabetes mellitus patients. The combination of insulin resistance and endothelial dysfunction leads to coronary artery disease and ischemic heart disease complications. A recognized biological marker, high-sensitivity C-reactive protein, has been used widely to assess the progression of atherosclerosis and inflammation. Along with coronary arterial damage and inflammatory processes, high-sensitivity C-reactive protein is considered as an essential atherosclerosis marker in patients with cardiovascular disease, but not as an insulin resistance marker in type 2 diabetes mellitus patients. A new biological marker that can act as a reliable indicator of both the exact state of insulin resistance and atherosclerosis is required to facilitate optimal health management of diabetic patients. Malfunctioning of insulin mechanism and endothelial dysfunction leads to innate immune activation and released several biological markers into circulation. This review examines potential biological markers, YKL-40, alpha-hydroxybutyrate, soluble CD36, leptin, resistin, interleukin-18, retinol binding protein-4, and chemerin, as they may play significant roles in insulin resistance and atherosclerosis in type 2 diabetes mellitus patients with coronary artery disease.
Collapse
Affiliation(s)
| | - Hasniza Zaman Huri
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Clinical Investigation Centre, 13th Floor Main Tower, University Malaya Medical Centre, 59100 Lembah Pantai, Kuala Lumpur, Malaysia
- *Hasniza Zaman Huri:
| | - Shireene Ratna Vethakkan
- Endocrinology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Wan Azman Wan Ahmad
- Clinical Investigation Centre, 13th Floor Main Tower, University Malaya Medical Centre, 59100 Lembah Pantai, Kuala Lumpur, Malaysia
- Cardiology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
86
|
Zamora C, Cantó E, Nieto JC, Angels Ortiz M, Juarez C, Vidal S. Functional consequences of CD36 downregulation by TLR signals. Cytokine 2012; 60:257-265. [PMID: 22795952 DOI: 10.1016/j.cyto.2012.06.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 12/20/2022]
Abstract
TLR recognition activates the secretion of pro- and anti-inflammatory cytokines and it also modulates the expression of crucial molecules involved in phagocytosis and antimicrobial activity. Scavenger receptors can act as TLR co-receptors or facilitate antigen loading. However, it remains unknown whether TLR can modulate the expression of these scavenger receptors. We stimulated human peripheral blood mononuclear cells (PBMC) with TLR2 (Pam3CSK4 and FSL1) and TLR4 ligand lipopolysaccharide (LPS) and then analyzed CD36 expression on different monocyte subpopulations by flow cytometry. TLR2 and TLR4 ligands can downregulate CD36 on the surface of monocytes, guiding the protein to intracellular compartments. Even though TLR-activation induced TNFα, IL-10 and IL-6 production, only recombinant TNFα was able to downregulate CD36. Neutralizing anti-TNFα antibodies showed that the Pam3CSK4 and FSL1-induced downregulation was partially mediated by TNFα but not by IL-6 or IL-10. However, LPS-induced downregulation could have also been caused by direct TLR4 targeting and signaling, and/or mediated by other unknown factors. CD36 downregulation reduced the capability of monocytes to phagocyte apoptotic neutrophils. In conclusion, modulation of scavenger receptor expression by TLR targeting on monocytes has functional consequences. Characterization this complex regulation may help us to understand this innate response and develop specific therapeutic drugs for each mechanism.
Collapse
Affiliation(s)
- Carlos Zamora
- Department of Immunology, Institut Recerca Hospital S. Pau, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
87
|
Liani R, Halvorsen B, Sestili S, Handberg A, Santilli F, Vazzana N, Formoso G, Aukrust P, Davì G. Plasma levels of soluble CD36, platelet activation, inflammation, and oxidative stress are increased in type 2 diabetic patients. Free Radic Biol Med 2012; 52:1318-24. [PMID: 22343420 DOI: 10.1016/j.freeradbiomed.2012.02.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/31/2012] [Accepted: 02/05/2012] [Indexed: 10/28/2022]
Abstract
Inflammation, oxidative stress, and platelet activation are involved in type 2 diabetes and its complications. Soluble CD36 (sCD36) has been proposed to early identify diabetics at risk of accelerated atherothrombosis. We aimed at characterizing the platelet contribution to sCD36 in diabetes, by correlating its concentration with the extent of platelet-mediated inflammation and in vivo lipid peroxidation and investigating the effects of low-dose aspirin on these processes. A cross-sectional comparison of sCD36, soluble CD40L (sCD40L) reflecting platelet-mediated inflammation, urinary 11-dehydro-TxB(2), and 8-iso-PGF(2α), in vivo markers of platelet activation and lipid peroxidation, was performed among 200 diabetic patients (94 of them on aspirin 100mg/day) and 47 healthy controls. sCD36 levels (median [IQR]: 0.72 [0.31-1.47] vs 0.26 [0.2-0.37], P=0.003) and urinary 11-dehydro-TxB(2) levels (666 [293-1336] vs 279 [160-396], P≤0.0001) were significantly higher in diabetic patients not on aspirin (n=106) than in healthy subjects. These variables were significantly lower in aspirin-treated diabetics than untreated patients (P<0.0001). Among patients not on aspirin, those with long-standing diabetes (>1 year) had significantly higher sCD36 levels in comparison to patients with diabetes duration <1 year (1.01 [0.62-1.86] vs 0.44 [0.22-1.21], P=0.001). sCD36 linearly correlated with sCD40L (rho=0.447; P=0.0001). On multiple regression analysis, 11-dehydro-TxB(2) (β=0.360; SEM=0.0001, P=0.001), 8-iso-PGF(2α) (β=0.469; SEM=0.0001, P<0.0001), and diabetes duration (β=0.244; SEM=0.207, P=0.017) independently predicted sCD36 levels. sCD36, platelet activation, inflammation, and oxidative stress are increased in type 2 diabetes. Future studies are needed to elucidate if the incomplete down-regulation of sCD36 by low-dose aspirin implies that sCD36 may be derived from tissues other than platelets or if additional antiplatelet strategies in diabetes are necessary to interrupt CD36-dependent platelet activation.
Collapse
Affiliation(s)
- Rossella Liani
- Center of Excellence on Aging, University of Chieti G. d'Annunzio, Via Colle dell'Ara, 66013, Chieti, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Qin B, Anderson RA, Kuzuya T, Kitaura Y, Shimomura Y. Multiple factors and pathways involved in hepatic very low density lipoprotein-apoB100 overproduction in Otsuka Long-Evans Tokushima Fatty rats. Atherosclerosis 2012; 222:409-16. [PMID: 22546076 DOI: 10.1016/j.atherosclerosis.2012.03.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 03/02/2012] [Accepted: 03/29/2012] [Indexed: 01/04/2023]
Abstract
AIMS Overproduction of hepatic very low-density lipoprotein (VLDL) particles is a major abnormality of lipoprotein dysregulation in type 2 diabetes (T2D). We sought to examine the relationship between systemic/hepatic inflammation associated with insulin resistance and apolipoprotein (apo)B100-containing VLDL production. METHODS AND RESULTS At the age of 19 wks, Otsuka Long-Evans Tokushima Fatty (OLETF) rats showed systemic inflammation (plasma TNF-α and interleukin (IL)-6 levels increased), insulin resistance (plasma retinol binding protein 4 and soluble CD36 levels were higher), dyslipidemia and fatty liver (plasma and liver triglyceride and cholesterol levels were higher as well as total VLDL-, VLDL(1)-, VLDL(2)-apoB100 and VLDL-triglycerides were overproduced), compared with the control rats. In livers of OLETF rats, mRNA levels of tnf, il1b and il6 were increased, but an anti-inflammatory protein, zinc finger protein 36, and its mRNA expression were decreased. We also found that the liver mRNA, protein levels, and tyrosine phosphorylation (pY) of insulin receptor (InsR) substrate (IRS) 2, but not IRS1, were decreased in OLETF rats; pY of InsR and Akt protein and phospho-Akt (ser437) were also reduced; but protein tyrosine phosphatase-1B protein was overexpressed. The gene expressions of glucose transporters 1 and 2, and glycogen synthase were decreased, but phosphatase and tensin homolog deleted on chromosome ten and glycogen synthase kinase 3β mRNAs were overexpressed, compared with the controls. Sterol regulatory element binding protein-1c mRNA, ATP-binding cassette transporter A1 mRNA, microsomal triglyceride transfer protein mRNA/protein, and CD36 mRNA/protein levels were increased and lipoprotein lipase and Niemann-Pick c1-like1 mRNA levels were decreased, which are all involved in lipogenesis. Decreased sirtuins1-3 mRNA levels were also observed in OLETF rats. CONCLUSIONS These abnormal genes, proteins expression and phosphorylation of multiple pathways related to inflammatory, insulin signaling and lipogenesis may be important underlying factors in VLDL-apoB100 particles overproduction observed in T2D. Our data contribute to the further understanding of an association of dyslipoproteinemia with systemic metabolic disorders, fatty liver and dysregulated hepatic metabolic pathways.
Collapse
Affiliation(s)
- Bolin Qin
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD 20705, USA.
| | | | | | | | | |
Collapse
|
89
|
Handberg A, Højlund K, Gastaldelli A, Flyvbjerg A, Dekker JM, Petrie J, Piatti P, Beck-Nielsen H. Plasma sCD36 is associated with markers of atherosclerosis, insulin resistance and fatty liver in a nondiabetic healthy population. J Intern Med 2012; 271:294-304. [PMID: 21883535 DOI: 10.1111/j.1365-2796.2011.02442.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Insulin resistance is associated with increased CD36 expression in a number of tissues. Moreover, excess macrophage CD36 may initiate atherosclerotic lesions. The aim of this study was to determine whether plasma soluble CD36 (sCD36) was associated with insulin resistance, fatty liver and carotid atherosclerosis in nondiabetic subjects. METHODS In 1296 healthy subjects without diabetes or hypertension recruited from 19 centres in 14 European countries (RISC study), we determined the levels of sCD36, adiponectin, lipids and liver enzymes, insulin sensitivity (M/I) by euglycaemic-hyperinsulinaemic clamp, carotid atherosclerosis as intima-media thickness (IMT) and two estimates of fatty liver, the fatty liver index (FLI) and liver fat percentage (LF%). RESULTS IMT, FLI, LF%, presence of the metabolic syndrome, impaired glucose regulation, insulin and triglycerides increased across sCD36 quartiles (Q2-Q4), whereas adiponectin and M/I decreased (P ≤ 0.01). sCD36 was lower in women than in men (P = 0.045). Log sCD36 showed a bimodal distribution, and amongst subjects with sCD36 within the log-normal distribution (log-normal population, n = 1029), sCD36 was increased in subjects with impaired glucose regulation (P = 0.045), metabolic syndrome (P = 0.006) or increased likelihood of fatty liver (P < 0.001). sCD36 correlated significantly with insulin, triglycerides, M/I and FLI (P < 0.05) after adjustment for study centre, gender, age, glucose tolerance status, smoking habits and alcohol consumption. In the log-normal population, these relationships were stronger than in the total study population and, additionally, sCD36 was significantly associated with LF% and IMT (P < 0.05). CONCLUSIONS In this cross-sectional study of nondiabetic subjects, sCD36 was significantly associated with indices of insulin resistance, carotid atherosclerosis and fatty liver. Prospective studies are needed to further evaluate the role of sCD36 in the inter-relationship between atherosclerosis, fatty liver and insulin resistance.
Collapse
Affiliation(s)
- A Handberg
- Department of Clinical Biochemistry, Aarhus Hospital and Aalborg Hospital, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Abstract
PURPOSE OF REVIEW The review summarizes our current understanding of the function of the fatty acid translocase, CD36, in lipid metabolism with an emphasis on the influence of CD36 genetic variants and their potential contribution to obesity-related complications. RECENT FINDINGS Studies in rodents implicate CD36 in a number of metabolic pathways with relevance to obesity and its associated complications. These include pathways related to fat utilization such as taste perception, intake, intestinal processing, and storage in adipose tissue. Dysfunction in these pathways, coupled with the ability of CD36 to transduce intracellular signals that initiate inflammation in response to excess fat supply, promotes metabolic pathology. In the last few years, the relevance of discoveries in rodents to humans has been highlighted by genetic studies, which identified common CD36 variants that influence circulating lipid levels and cardiometabolic phenotypes. SUMMARY Recent genetic studies suggest that CD36 plays an important role in lipid metabolism in humans and may be involved in obesity-related complications. These findings may accelerate the translation of CD36 metabolic functions determined in rodents to humans. Importantly, these studies highlight the potential utility of assessing CD36 expression and common single-nucleotide polymorphism genotypes.
Collapse
|
91
|
Kuda O, Jenkins CM, Skinner JR, Moon SH, Su X, Gross RW, Abumrad NA. CD36 protein is involved in store-operated calcium flux, phospholipase A2 activation, and production of prostaglandin E2. J Biol Chem 2011; 286:17785-95. [PMID: 21454644 PMCID: PMC3093854 DOI: 10.1074/jbc.m111.232975] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 03/29/2011] [Indexed: 12/20/2022] Open
Abstract
The scavenger receptor FAT/CD36 contributes to the inflammation associated with diabetes, atherosclerosis, thrombosis, and Alzheimer disease. Underlying mechanisms include CD36 promotion of oxidative stress and its signaling to stress kinases. Here we document an additional mechanism for the role of CD36 in inflammation. CD36 regulates membrane calcium influx in response to endoplasmic reticulum (ER) stress, release of arachidonic acid (AA) from cellular membranes by cytoplasmic phospholipase A(2)α (cPLA(2)α) and contributes to the generation of proinflammatory eicosanoids. CHO cells stably expressing human CD36 released severalfold more AA and prostaglandin E(2) (PGE(2)), a major product of AA metabolism by cyclooxygenases, in response to thapsigargin-induced ER stress as compared with control cells. Calcium influx after ER calcium release resulted in phosphorylation of cPLA(2) and its translocation to membranes in a CD36-dependent manner. Peritoneal macrophages from CD36(-/-) mice exhibited diminished calcium transients and reduced AA release after thapsigargin or UTP treatment with decreased ERK1/2 and cPLA(2) phosphorylation. However, PGE(2) production was unexpectedly enhanced in CD36(-/-) macrophages, which probably resulted from a large induction of cyclooxygenase 2 mRNA and protein. The data demonstrate participation of CD36 in membrane calcium influx in response to ER stress or purinergic receptor stimulation resulting in AA liberation for PGE(2) formation. Collectively, these results identify a mechanism contributing to the pleiotropic proinflammatory effects of CD36 and suggest that its targeted inhibition may reduce the acute inflammatory response.
Collapse
Affiliation(s)
- Ondrej Kuda
- From the Department of Medicine, Center for Human Nutrition, and
| | - Christopher M. Jenkins
- the Division of Bioorganic Chemistry and Molecular Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - James R. Skinner
- From the Department of Medicine, Center for Human Nutrition, and
| | - Sung Ho Moon
- the Division of Bioorganic Chemistry and Molecular Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Xiong Su
- From the Department of Medicine, Center for Human Nutrition, and
| | - Richard W. Gross
- the Division of Bioorganic Chemistry and Molecular Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Nada A. Abumrad
- From the Department of Medicine, Center for Human Nutrition, and
| |
Collapse
|
92
|
Koonen DPY, Jensen MK, Handberg A. Soluble CD36- a marker of the (pathophysiological) role of CD36 in the metabolic syndrome? Arch Physiol Biochem 2011; 117:57-63. [PMID: 21250778 DOI: 10.3109/13813455.2010.543136] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CD36 is a class B scavenger receptor observed in many cell types and tissues throughout the body. Recent literature has implicated CD36 in the pathogenesis of metabolic dysregulation such as found in obesity, insulin resistance, and atherosclerosis. Genetic variation at the CD36 loci have been associated with obesity and lipid components of the metabolic syndrome, with risk of heart disease and type 2 diabetes. Recently, non-cell bound CD36 was identified in human plasma and was termed soluble CD36 (sCD36). In this review we will describe the functions of CD36 in tissues and address the role of sCD36 in the context of the metabolic syndrome. We will also highlight recent findings from human genetic studies looking at the CD36 locus in relation to metabolic profile in the general population. Finally, we present a model in which insulin resistance, oxLDL, low-grade inflammation and liver steatosis may contribute to elevated levels of sCD36.
Collapse
Affiliation(s)
- Debby P Y Koonen
- Molecular Genetics, Department of Pathology and Medical Biology, University Medical Center Groningen, The Netherlands
| | | | | |
Collapse
|
93
|
Carotid plaque age is a feature of plaque stability inversely related to levels of plasma insulin. PLoS One 2011; 6:e18248. [PMID: 21490968 PMCID: PMC3072386 DOI: 10.1371/journal.pone.0018248] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 03/02/2011] [Indexed: 12/25/2022] Open
Abstract
Background The stability of atherosclerotic plaques determines the risk for rupture,
which may lead to thrombus formation and potentially severe clinical
complications such as myocardial infarction and stroke. Although the rate of
plaque formation may be important for plaque stability, this process is not
well understood. We took advantage of the atmospheric
14C-declination curve (a result of the atomic bomb tests in the
1950s and 1960s) to determine the average biological age of carotid
plaques. Methodology/Principal Finding The cores of carotid plaques were dissected from 29 well-characterized,
symptomatic patients with carotid stenosis and analyzed for 14C
content by accelerator mass spectrometry. The average plaque age (i.e.
formation time) was 9.6±3.3 years. All but two plaques had formed
within 5–15 years before surgery. Plaque age was not associated with
the chronological ages of the patients but was inversely related to plasma
insulin levels (p = 0.0014). Most plaques were
echo-lucent rather than echo-rich (2.24±0.97, range 1–5).
However, plaques in the lowest tercile of plaque age (most recently formed)
were characterized by further instability with a higher content of lipids
and macrophages (67.8±12.4 vs. 50.4±6.2,
p = 0.00005; 57.6±26.1 vs. 39.8±25.7,
p<0.0005, respectively), less collagen (45.3±6.1 vs.
51.1±9.8, p<0.05), and fewer smooth muscle cells (130±31
vs. 141±21, p<0.05) than plaques in the highest tercile.
Microarray analysis of plaques in the lowest tercile also showed increased
activity of genes involved in immune responses and oxidative
phosphorylation. Conclusions/Significance Our results show, for the first time, that plaque age, as judge by relative
incorporation of 14C, can improve our understanding of carotid
plaque stability and therefore risk for clinical complications. Our results
also suggest that levels of plasma insulin might be involved in determining
carotid plaque age.
Collapse
|
94
|
Alkhatatbeh MJ, Mhaidat NM, Enjeti AK, Lincz LF, Thorne RF. The putative diabetic plasma marker, soluble CD36, is non-cleaved, non-soluble and entirely associated with microparticles. J Thromb Haemost 2011; 9:844-51. [PMID: 21276198 DOI: 10.1111/j.1538-7836.2011.04220.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND CD36 is a widely expressed cell surface receptor that binds lipoproteins, and its function has been implicated in many complications of the metabolic syndrome. A cell-free form of CD36, soluble CD36 (sCD36), has been reported in human plasma, found to be elevated in obesity and diabetes, and claimed as a marker of insulin resistance. OBJECTIVE To determine the nature of sCD36; in particular, whether sCD36 is truly soluble or, as hypothesized, is found as a component of circulating microparticles (MPs). METHODS Lipoproteins were fractionated by density gradient centrifugation, and plasma MPs were isolated by ultracentrifugation, size exclusion, and immunoprecipitation with CD36 detected by immunoblotting. MPs from plasma and activated platelets were analyzed by multicolor flow cytometry, with a DyLight-488 anti-CD36 conjugate in combination with antibodies against different cellular markers. RESULTS Cell-free plasma CD36 was not observed associated with lipoproteins and was not a proteolytic fragment; rather, it was associated with the plasma MP fraction, suggesting that sCD36 in the plasma of normal subjects is a product of circulating MPs. Cytometric and immunoblotting analyses of plasma from normal donors showed that these MPs were derived mainly from platelets. Analysis of in vitro activated platelets also showed that CD36 to be secreted in the form of MPs. CONCLUSIONS sCD36 is not a proteolytic product, but rather is associated with a specific subset of circulating MPs that can readily be analysed. This finding will enable more specific investigations into the cellular source of the increased levels of plasma CD36 found in subjects with diabetes.
Collapse
Affiliation(s)
- M J Alkhatatbeh
- Cancer Research Unit, School of Biomedical Sciences and Pharmacy, Faculty of Health, the University of Newcastle, NSW, Australia
| | | | | | | | | |
Collapse
|
95
|
Kennedy DJ, Kashyap SR. Pathogenic role of scavenger receptor CD36 in the metabolic syndrome and diabetes. Metab Syndr Relat Disord 2011; 9:239-45. [PMID: 21428745 DOI: 10.1089/met.2011.0003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Obesity is increasing at epidemic proportions in the United States and is a major contributor to the development of both metabolic syndrome (glucose intolerance, dyslipidemia, hypertension) and atherosclerotic cardiovascular disease. A wide body of evidence has linked systemic low-grade inflammation as underlying obesity and insulin-resistant states via monocyte/macrophage activation. Transgenic deletion of scavenger receptor type B CD36 in rodents has suggested a pivotal role for CD36 in mediating inflammation, insulin resistance, and atherogenesis through transport of fatty acids and uptake of oxidized lipids, respectively. CD36 signaling pathways involving c-Jun N-terminal kinase (JNK) activation and Toll-like receptors have been implicated in the induction of insulin resistance. This review will focus on the pathogenic role of CD36 receptors in metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- David J Kennedy
- Department of Cell Biology, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Ohio 44195, USA
| | | |
Collapse
|
96
|
Banerjee M, Gautam S, Saxena M, Bid HK, Agrawal C. Association of CD36 gene variants rs1761667 (G>A) and rs1527483 (C>T) with Type 2 diabetes in North Indian population. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.ijdm.2010.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
97
|
Allred CC, Krennmayr T, Koutsari C, Zhou L, Ali AH, Jensen MD. A novel ELISA for measuring CD36 protein in human adipose tissue. J Lipid Res 2010; 52:408-15. [PMID: 21115967 DOI: 10.1194/jlr.m008995] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
CD36 is a transmembrane protein present in many tissues that is believed to facilitate inward fatty acid transport. Western blotting is the most widely used method to measure tissue CD36 protein content, but it is time consuming, technically demanding, and semiquantitative. To more precisely measure adipose tissue CD36 content we developed an enzyme linked immunosorbent assay (ELISA) after establishing that: 1) the anti-CD36 antibodies gave a single distinct band on traditional Western blots, and 2) the vast majority of adipocyte CD36 resides in the plasma membrane. By using serial dilutions of each sample and including a calibrator sample and quality control sample on each plate, we could achieve inter- and intra-assay variability of ∼ 10%. We found that CD36 content in omental and abdominal subcutaneous adipose tissue varied over a 2-5-fold range depending upon the means of data expression (per units of tissue protein, weight, or lipid). Omental CD36 content in women decreased markedly (P = 0.01) as a function of fat cell size. For the most part, tissue CD36 content was not correlated with CD36 mRNA. This ELISA method for tissue CD36 content should enhance research into the role of this protein on tissue fatty acid uptake.
Collapse
Affiliation(s)
- Carolyn C Allred
- Endocrine Research Unit, Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | |
Collapse
|
98
|
Love-Gregory L, Sherva R, Schappe T, Qi JS, McCrea J, Klein S, Connelly MA, Abumrad NA. Common CD36 SNPs reduce protein expression and may contribute to a protective atherogenic profile. Hum Mol Genet 2010; 20:193-201. [PMID: 20935172 DOI: 10.1093/hmg/ddq449] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Membrane CD36 functions in the uptake of fatty acids (FAs), oxidized lipoproteins and in signal transduction after binding these ligands. In rodents, CD36 is implicated in abnormal lipid metabolism, inflammation and atherosclerosis. In humans, CD36 variants have been identified to influence free FA and high-density lipoprotein (HDL) levels and to associate with the risk of the metabolic syndrome, coronary artery disease and stroke. In this study, 15 common lipid-associated CD36 single nucleotide polymorphisms (SNPs) were evaluated for the impact on monocyte CD36 expression (protein and transcript) in 104 African Americans. In a subset of subjects, the SNPs were tested for association with monocyte surface CD36 (n=65) and platelet total CD36 (n=57). The relationship between CD36 expression and serum HDL and very low-density lipoproteins (VLDLs) levels was also examined. After a permutation-based correction for multiple tests, four SNPs (rs1761667, rs3211909, rs3211913, rs3211938) influenced monocyte CD36 protein and two (rs3211909, rs3211938) platelet CD36. The effect of the HDL-associated SNPs on CD36 expression inversely related to the impact on serum HDL and potential causality was supported by Mendelian randomization analysis. Consistent with this, monocyte CD36 protein negatively correlated with total HDL and HDL subfractions. In contrast, positive correlations were documented between monocyte CD36 and VLDL lipid, particle number and apolipoprotein B. In conclusion, CD36 variants that reduce protein expression appear to promote a protective metabolic profile. The SNPs in this study may have predictive potential on CD36 expression and disease susceptibility in African Americans. Further studies are warranted to validate and determine whether these findings are population specific.
Collapse
Affiliation(s)
- Latisha Love-Gregory
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Serum soluble CD36, assessed by a novel monoclonal antibody-based sandwich ELISA, predicts cardiovascular mortality in dialysis patients. Clin Chim Acta 2010; 411:2079-82. [PMID: 20837002 DOI: 10.1016/j.cca.2010.09.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 08/12/2010] [Accepted: 09/06/2010] [Indexed: 12/25/2022]
Abstract
AIM Accelerated atherosclerosis is a characteristic feature of chronic kidney disease (CKD). CD36 is a scavenger receptor which contributes to foam cell formation, an early crucial step in atherosclerosis development. Recently, a soluble form of CD36 (sCD36) has been discovered. The aim of the study was to develop an ELISA method for quantitative sCD36 evaluation and to measure it in a cohort of CKD stage 5 patients. METHOD A novel monoclonal antibody-based sandwich ELISA for sCD36 evaluation was developed and verified by repeated optimization procedures. Serum concentration of sCD36 was then analyzed in a cohort of 228 CKD stage 5 patients prior to dialysis initiation. Additionally, samples from a control group of 73 healthy, age and gender-matched subjects were evaluated. RESULTS The novel CD36 ELISA assay had a recovery of at least 90%, and intra- and inter-assay variability of 6 and 11%, respectively. Concentration of serum sCD36 in CKD patients was significantly increased as compared to controls, and associated with the use of HMG-CoA reductase inhibitors (statins) and the presence of diabetes mellitus (DM). Patients above the 75th percentile of sCD36 concentration were at increased risk of 3-year cardiovascular mortality, as compared to the rest of the cohort [HR 2.85 (1.09-7.59) p=0.03]. CONCLUSION For the first time, sCD36 was assessed quantitatively in a group of patients and showed associations with DM, CKD, and statin use. Furthermore, the concentration of sCD36 predicted cardiovascular mortality in CKD stage 5 patients.
Collapse
|
100
|
Qin B, Polansky MM, Harry D, Anderson RA. Green tea polyphenols improve cardiac muscle mRNA and protein levels of signal pathways related to insulin and lipid metabolism and inflammation in insulin-resistant rats. Mol Nutr Food Res 2010; 54 Suppl 1:S14-23. [PMID: 20112301 DOI: 10.1002/mnfr.200900306] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Epidemiological studies indicate that the consumption of green tea polyphenols (GTP) may reduce the risk of coronary artery disease. To explore the underlying mechanisms of action at the molecular level, we examined the effects of GTP on the cardiac mRNA and protein levels of genes involved in insulin and lipid metabolism and inflammation. In rats fed a high-fructose diet, supplementation with GTP (200 mg/kg BW daily dissolved in distilled water) for 6 wk, reduced systemic blood glucose, plasma insulin, retinol-binding protein 4, soluble CD36, cholesterol, triglycerides, free fatty acids and LDL-C levels, as well as the pro-inflammatory cytokines, tumor necrosis factor-alpha (TNF-alpha) and IL-6. GTP did not affect food intake, bodyweight and heart weight. In the myocardium, GTP also increased the insulin receptor (Ir), insulin receptor substrate 1 and 2 (Irs1 and Irs2), phosphoinositide-3-kinase (Pi3k), v-akt murine thymoma viral oncogene homolog 1 (Akt1), glucose transporter 1 and 4 (Glut1 and Glut4) and glycogen synthase 1 (Gys1) expression but inhibited phosphatase and tensin homolog deleted on chromosome ten (Pten) expression and decreased glycogen synthase kinase 3beta (Gsk3beta) mRNA expression. The sterol regulatory element-binding protein-1c (Srebp1c) mRNA, microsomal triglyceride transfer protein (Mttp) mRNA and protein, Cd36 mRNA and cluster of differentiation 36 protein levels were decreased and peroxisome proliferator-activated receptor (Ppar)gamma mRNA levels were increased. GTP also decreased the inflammatory factors: Tnf, Il1b and Il6 mRNA levels, and enhanced the anti-inflammatory protein, zinc-finger protein, protein and mRNA expression. In summary, consumption of GTP ameliorated the detrimental effects of high-fructose diet on insulin signaling, lipid metabolism and inflammation in the cardiac muscle of rats.
Collapse
Affiliation(s)
- Bolin Qin
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Beltsville, MD 20705, USA
| | | | | | | |
Collapse
|