1
|
van der Ark-Vonk EM, Puijk MV, Pasterkamp G, van der Laan SW. The Effects of FABP4 on Cardiovascular Disease in the Aging Population. Curr Atheroscler Rep 2024; 26:163-175. [PMID: 38698167 PMCID: PMC11087245 DOI: 10.1007/s11883-024-01196-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 05/05/2024]
Abstract
PURPOSE OF REVIEW Fatty acid-binding protein 4 (FABP4) plays a role in lipid metabolism and cardiovascular health. In this paper, we cover FABP4 biology, its implications in atherosclerosis from observational studies, genetic factors affecting FABP4 serum levels, and ongoing drug development to target FABP4 and offer insights into future FABP4 research. RECENT FINDINGS FABP4 impacts cells through JAK2/STAT2 and c-kit pathways, increasing inflammatory and adhesion-related proteins. In addition, FABP4 induces angiogenesis and vascular smooth muscle cell proliferation and migration. FABP4 is established as a reliable predictive biomarker for cardiovascular disease in specific at-risk groups. Genetic studies robustly link PPARG and FABP4 variants to FABP4 serum levels. Considering the potential effects on atherosclerotic lesion development, drug discovery programs have been initiated in search for potent inhibitors of FABP4. Elevated FABP4 levels indicate an increased cardiovascular risk and is causally related to acceleration of atherosclerotic disease, However, clinical trials for FABP4 inhibition are lacking, possibly due to concerns about available compounds' side effects. Further research on FABP4 genetics and its putative causal role in cardiovascular disease is needed, particularly in aging subgroups.
Collapse
Affiliation(s)
- Ellen M van der Ark-Vonk
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Mike V Puijk
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Sander W van der Laan
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
2
|
Liu Y, Zhao Y, Guo Z, Li M, Shan H, Zhang Y, Miao C, Gu Y. Pericarotid Fat Stranding at Computed Tomography Angiography: A Marker of the Short-Term Prognosis of Acute Ischemic Stroke. J Comput Assist Tomogr 2024; 48:311-316. [PMID: 37876252 DOI: 10.1097/rct.0000000000001555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
PURPOSE Perivascular epicardial fat stranding detected in the coronary computed tomography (CT) angiography is associated with culprit lesions and provides helpful information on the risk of acute coronary syndrome. This study aimed to evaluate the potential clinical significance of pericarotid fat stranding (PCFS) and investigate the association between PCFS and short-term prognosis in acute stroke using head and neck CT angiography (CTA). METHODS This study included 80 patients (mean age, 69.69 ± 11.03; 58 men) who underwent both head and neck CTA and magnetic resonance imaging within a 1-week period. Baseline characteristics, pericarotid adipose tissue attenuation, plaque characteristics, ischemic penumbra, infarct core volume, infarct core growth rate (CGR), and the grade of collateral status were recorded and compared between a PCFS group and a non-PCFS group. Data were compared using the 2-sample t test, Mann-Whitney U test, Fisher exact test, and Spearman rank correlation analysis. RESULTS We found that patients with PCFS had a significantly higher pericarotid adipose tissue density than patients without PCFS (-55.75 ± 5.53 vs -65.82 ± 9.65, P < 0.001). Patients with PCFS showed a larger infarct core volume (166.43 ± 73.07 vs 91.43 ± 55.03, P = 0.001) and faster CGR (39.57 ± 12.01 vs 19.83 ± 32.77; P < 0.001), and the frequency of adverse prognosis was more significant than in control participants (83.33% vs 19.11%). CONCLUSIONS Individuals with PCFS showed higher CGR, which was substantially related to worse outcomes in patients with acute stroke with ipsilateral carotid atherosclerosis. Recognition of PCFS may help predict stroke prognosis and allow doctors to take early action to improve patient prognosis.
Collapse
Affiliation(s)
- Ying Liu
- From the Department of Radiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Jalilian N, Pakzad R, Shahbazi M, Edrisi SR, Haghani K, Jalilian M, Bakhtiyari S. Circulating FABP-4 Levels in Patients with Atherosclerosis or Coronary Artery Disease: A Comprehensive Systematic Review and Meta-Analysis. Cardiovasc Ther 2023; 2023:1092263. [PMID: 38024104 PMCID: PMC10673666 DOI: 10.1155/2023/1092263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 10/22/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Background Cardiovascular diseases (CDs), notably coronary artery disease (CAD) due to atherosclerosis, impose substantial global health and economic burdens. Fatty acid-binding proteins (FABPs), including FABP-4, have been recently linked to CDs. This study conducted a systematic review and meta-analysis to examine FABP-4 levels in CAD and atherosclerosis patients, exploring their potential links to these conditions. Methods A systematic review and meta-analysis were done based on the PRISMA guideline. The international databases including Medline, Embase, Cochrane Library, Scopus, Web of Science, and UpToDate were searched to find all related studies on the effect of FABP-4 on patients with CAD or atherosclerosis which were published till June 2022 without language restriction. The Cochran's Q-test and I2 statistic were applied to assess heterogeneity, a random effect model was used to estimate the pooled standardized mean difference (SMD), a metaregression method was utilized to investigate the factors affecting heterogeneity between studies, and Egger's test was used to assess the publication bias. Results Of 1051 studies, 9 studies with a sample size of 2327 were included in the systematic review and meta-analysis. The level of circulating FABP-4 in the patient groups was significantly higher than in the control groups (SMD = 0.60 (95% CI: 0.30 to 0.91, I2: 91.47%)). The SMD in female and male patients were 0.26 (95% CI: 0.01 to 0.52, I2: 0%) and 0.22 (95% CI: 0.08 to 0.35, I2: 44.7%), respectively. There was considerable heterogeneity between the studies. The countries had a positive relationship with heterogeneity (coefficient = 0.29, p < 0.001); but BMI, lipid indices, gender, study design, and type of kit had no effect on the heterogeneity. No publication bias was observed (p: 0.137). Conclusion In summary, this meta-analysis revealed elevated circulating FABP-4 levels in CDs, suggesting its potential as a biomarker for these conditions. Further research is warranted to explore its clinical relevance.
Collapse
Affiliation(s)
- Narges Jalilian
- Department of Medical Biochemistry, Faculty of Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Reza Pakzad
- Student Research Committee, Ilam University of Medical Sciences, Ilam, Iran
- Department of Epidemiology, Faculty of Health, Ilam University of Medical Sciences, Ilam, Iran
- Health and Environment Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Mahdi Shahbazi
- Department of Medical Biochemistry, Faculty of Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Seyyed-Reza Edrisi
- Department of Medical Biochemistry, Faculty of Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Karimeh Haghani
- Department of Medical Biochemistry, Faculty of Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Mohsen Jalilian
- Department of Health Education and Promotion, Faculty of Health, Ilam University of Medical Sciences, Ilam, Iran
| | - Salar Bakhtiyari
- Department of Medical Biochemistry, Faculty of Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, USA
| |
Collapse
|
4
|
Zhou S, Wang X, Han Q, He L, Tang W, Zhang A. Elevated serum fatty acid-binding protein 4 level predicts all-cause and cardiovascular mortality in peritoneal dialysis patients: a five-year study. Ren Fail 2023; 45:2262624. [PMID: 37782286 PMCID: PMC10547445 DOI: 10.1080/0886022x.2023.2262624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND To explore the prospective role of serum fatty acid-binding protein 4 (FABP4) in the outcomes of peritoneal dialysis (PD) patients. METHODS A prospective observational study was conducted with 159 patients on PD. Demographic and clinical data at baseline were collected from medical records. Biochemical data were recorded based on blood samples measured in a central laboratory. Serum FABP4 concentrations were determined using enzyme-linked immunosorbent assay. Body composition was measured using a Body Composition Monitor. Abdominal lateral plain radiography was used to evaluate vascular calcification. The primary endpoints were all-cause and cardiovascular death. RESULTS The median of serum FABP4 concentration was 154.6 ng/mL (interquartile range, 132.8-269.7 ng/mL). Increased serum FABP4 was associated with increased vascular calcification proportion, time on dialysis, body mass index, high-sensitivity C-reactive protein (hs-CRP), intact parathyroid hormone (iPTH), triglycerides, body fat mass, and body fat percentage (p < 0.05). Increased serum FABP4 was associated with decreased residual kidney Kt/V urea (p < 0.05). Patients with hs-CRP≥ 3 mg/L had significantly higher serum FABP4 than those with hs-CRP< 3 mg/L (p < 0.05). Patients with vascular calcification had significantly higher serum FABP4 than those without vascular calcification (p < 0.05). During a median follow-up of 58.0 months, 58 all-cause deaths and 26 cardiovascular deaths occurred. High serum FABP4 levels were independently predictive for all-cause [hazard ratio (HR), 1.003; 95% confidence interval (CI), 1.001-1.005; p = 0.016] and cardiovascular death (HR, 1.005; 95% CI, 1.001-1.008; p = 0.006) in PD patients. CONCLUSIONS Increased serum FABP4 levels can independently predict all-cause and cardiovascular death in patients on PD.
Collapse
Affiliation(s)
- Sijia Zhou
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Xiaoxiao Wang
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Qingfeng Han
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Lian He
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Wen Tang
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Aihua Zhang
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Lv J, Hu Y, Li L, He Y, Wang J, Guo N, Fang Y, Chen Q, Cai C, Tong J, Tang L, Wang Z. Targeting FABP4 in elderly mice rejuvenates liver metabolism and ameliorates aging-associated metabolic disorders. Metabolism 2023; 142:155528. [PMID: 36842611 DOI: 10.1016/j.metabol.2023.155528] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 02/28/2023]
Abstract
INTRODUCTION Aging is characterized by progressive metabolic dyshomeostasis that increases morbidity and mortality. Solutions for optimizing healthy aging are challenged by lacking appropriate biomarkers. Moreover, druggable targets to rejuvenate the aging-associated metabolic phenotypes remain unavailable. METHODS Proteomics analysis was performed in a cohort of young and elderly adults. Circulating levels of insulin-like growth factor 1 (IGF-1) and fatty acid binding protein 4 (FABP4) were evaluated by ELISA. FABP4 was silenced in elderly mice by adeno-associated virus. Metabolic activities were measured by metabolic cages. Cognitive function was evaluated by Morris water maze. Glucose and lipid metabolism were evaluated by biochemistry assays with blood samples. RNA-seq in mouse liver was performed for transcriptome analysis. RESULTS Among 9 aging-sensitive proteins shared by both male and female, FABP4 was identified as a reliable aging biomarker in both human and mouse. Silencing FABP4 in elderly mice significantly rejuvenated the aging-associated decline in metabolic activities. FABP4 knockdown reversed the aging-associated metabolic disorders by promoting degradation of cholesterol and fatty acids, while suppressing gluconeogenesis. Transcriptome analysis revealed a restoration of the pro-aging gene reprogramming towards inflammation and metabolic disorders in the liver after FABP4 knockdown. FABP4 overexpression promoted human LO2 cell senescence. Moreover, administration of an FABP4 inhibitor BMS309403 delivered metabolic benefits in elderly mice. CONCLUSION Our findings demonstrate FABP4 as a reliable aging biomarker as well as a practicable target to improve healthy aging in the elderly.
Collapse
Affiliation(s)
- Jian Lv
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yimeng Hu
- Department of Endocrinology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Lili Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuan He
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingjing Wang
- School of Martial Arts, Wuhan Sports University, Wuhan 430079, China
| | - Ningning Guo
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Fang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qin Chen
- School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Cheguo Cai
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Jingjing Tong
- School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| | - Lixu Tang
- School of Martial Arts, Wuhan Sports University, Wuhan 430079, China.
| | - Zhihua Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Hyder A. Naturally-occurring carboxylic acids from traditional antidiabetic plants as potential pancreatic islet FABP3 inhibitors. A molecular docking-aided study. Chem Biol Interact 2023; 372:110368. [PMID: 36709838 DOI: 10.1016/j.cbi.2023.110368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
The antidiabetic action of traditional plants is mostly attributed to their antioxidant and anti-inflammatory properties. These plants are still having some secrets, making them an attractive source that allows for investigating new drugs or uncovering precise pharmacologic antidiabetic functions of their constituents. In diabetes, which is a lipid disease, long-term exposure of pancreatic islet beta cells to fatty acids (FAs) increases basal insulin release, reduces glucose-stimulated insulin secretion, causes islet beta cell inflammation, failure and apoptosis. Pancreatic islet beta cells express fatty acid binding protein 3 (FABP3) that receives long-chain FAs and traffics them throughout different cellular compartments to be metabolized and render their effects. Inhibition of this FABP3 may retard FA metabolism and protect islet beta cells. Since FAs interact with FABPs by their carboxylic group, some traditionally-known antidiabetic plants were reviewed in the present study, searching for their components that have common features of FABP ligands, namely carboxylic group and hydrophobic tail. Many of these carboxylic acids were computationally introduced into the ligand-binding pocket of FABP3 and some of them exhibited FABP3 ligand possibilities. Among others, the naturally occurring ferulic, cleomaldeic, caffeic, sinapic, hydroxycinnamic, 4-p-coumaroylquinic, quinoline-2-carboxylic, chlorogenic, 6-hydroxykynurenic, and rosmarinic acids in many plants are promising candidates for being FABP3-specific inhibitors. The study shed light on repurposing these phyto-carboxylic acids to function as FABP inhibitors. However, more in-depth biological and pharmacological studies to broaden the understanding of this function are needed.
Collapse
Affiliation(s)
- Ayman Hyder
- Faculty of Science, Damietta University, New Damietta 34517, Egypt.
| |
Collapse
|
7
|
de Melo TC, Trevisan-Silva D, Alvarez-Flores MP, Gomes RN, de Souza MM, Valerio HP, Oliveira DS, DeOcesano-Pereira C, Botosso VF, Calil Jorge SA, Schattner M, Gomez RM, Chudzinski-Tavassi AM. Proteomic Analysis Identifies Molecular Players and Biological Processes Specific to SARS-CoV-2 Exposure in Endothelial Cells. Int J Mol Sci 2022; 23:10452. [PMID: 36142365 PMCID: PMC9500950 DOI: 10.3390/ijms231810452] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/21/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been responsible for the severe pandemic of acute respiratory disease, coronavirus disease 2019 (COVID-19), experienced in the 21st century. The clinical manifestations range from mild symptoms to abnormal blood coagulation and severe respiratory failure. In severe cases, COVID-19 manifests as a thromboinflammatory disease. Damage to the vascular compartment caused by SARS-CoV-2 has been linked to thrombosis, triggered by an enhanced immune response. The molecular mechanisms underlying endothelial activation have not been fully elucidated. We aimed to identify the proteins correlated to the molecular response of human umbilical vein endothelial cells (HUVECs) after exposure to SARS-CoV-2, which might help to unravel the molecular mechanisms of endothelium activation in COVID-19. In this direction, we exposed HUVECs to SARS-CoV-2 and analyzed the expression of specific cellular receptors, and changes in the proteome of HUVECs at different time points. We identified that HUVECs exhibit non-productive infection without cytopathic effects, in addition to the lack of expression of specific cell receptors known to be essential for SARS-CoV-2 entry into cells. We highlighted the enrichment of the protein SUMOylation pathway and the increase in SUMO2, which was confirmed by orthogonal assays. In conclusion, proteomic analysis revealed that the exposure to SARS-CoV-2 induced oxidative stress and changes in protein abundance and pathways enrichment that resembled endothelial dysfunction.
Collapse
Affiliation(s)
- Thatiana Corrêa de Melo
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Dilza Trevisan-Silva
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Miryam P. Alvarez-Flores
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Renata Nascimento Gomes
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Marcelo Medina de Souza
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Hellen Paula Valerio
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Douglas S. Oliveira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | - Carlos DeOcesano-Pereira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo 05503900, Brazil
| | | | | | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (IMEX-CONICET-ANM), Buenos Aires 1425, Argentina
| | - Ricardo M. Gomez
- Laboratory of Animal Viruses, Institute of Biotechnology and Molecular Biology, CONICET-UNLP, La Plata 1900, Argentina
| | | |
Collapse
|
8
|
Kigka VI, Potsika V, Mantzaris M, Tsakanikas V, Koncar I, Fotiadis DI. Serum Biomarkers in Carotid Artery Disease. Diagnostics (Basel) 2021; 11:diagnostics11112143. [PMID: 34829489 PMCID: PMC8619296 DOI: 10.3390/diagnostics11112143] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Carotid artery disease is considered a major cause of strokes and there is a need for early disease detection and management. Although imaging techniques have been developed for the diagnosis of carotid artery disease and different imaging-based markers have been proposed for the characterization of atherosclerotic plaques, there is still need for a definition of high-risk plaques in asymptomatic patients who may benefit from surgical intervention. Measurement of circulating biomarkers is a promising method to assist in patient-specific disease management, but the lack of robust clinical evidence limits their use as a standard of care. The purpose of this review paper is to present circulating biomarkers related to carotid artery diagnosis and prognosis, which are mainly provided by statistical-based clinical studies. The result of our investigation showed that typical well-established inflammatory biomarkers and biomarkers related to patient lipid profiles are associated with carotid artery disease. In addition to this, more specialized types of biomarkers, such as endothelial and cell adhesion, matrix degrading, and metabolic biomarkers seem to be associated with different carotid artery disease outputs, assisting vascular specialists in selecting patients at high risk for stroke and in need of intervention.
Collapse
Affiliation(s)
- Vassiliki I. Kigka
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece; (V.I.K.); (V.P.); (M.M.); (V.T.)
| | - Vassiliki Potsika
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece; (V.I.K.); (V.P.); (M.M.); (V.T.)
| | - Michalis Mantzaris
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece; (V.I.K.); (V.P.); (M.M.); (V.T.)
| | - Vassilis Tsakanikas
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece; (V.I.K.); (V.P.); (M.M.); (V.T.)
| | - Igor Koncar
- Department of Vascular and Endovascular Surgery, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Department of Vascular and Endovascular Surgery, Clinic Center of Serbia, 11000 Belgrade, Serbia
| | - Dimitrios I. Fotiadis
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece; (V.I.K.); (V.P.); (M.M.); (V.T.)
- Institute of Molecular Biology and Biotechnology, Department of Biomedical Research Institute—FORTH, University Campus of Ioannina, 45110 Ioannina, Greece
- Correspondence: ; Tel.: +30-26510-09006; Fax: +30-26510-08889
| |
Collapse
|
9
|
A-FABP in Metabolic Diseases and the Therapeutic Implications: An Update. Int J Mol Sci 2021; 22:ijms22179386. [PMID: 34502295 PMCID: PMC8456319 DOI: 10.3390/ijms22179386] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Adipocyte fatty acid-binding protein (A-FABP), which is also known as ap2 or FABP4, is a fatty acid chaperone that has been further defined as a fat-derived hormone. It regulates lipid homeostasis and is a key mediator of inflammation. Circulating levels of A-FABP are closely associated with metabolic syndrome and cardiometabolic diseases with imminent diagnostic and prognostic significance. Numerous animal studies have elucidated the potential underlying mechanisms involving A-FABP in these diseases. Recent studies demonstrated its physiological role in the regulation of adaptive thermogenesis and its pathological roles in ischemic stroke and liver fibrosis. Due to its implication in various diseases, A-FABP has become a promising target for the development of small molecule inhibitors and neutralizing antibodies for disease treatment. This review summarizes the clinical and animal findings of A-FABP in the pathogenesis of cardio-metabolic diseases in recent years. The underlying mechanism and its therapeutic implications are also highlighted.
Collapse
|
10
|
Li H, Xu X, Luo B, Zhang Y. The Predictive Value of Carotid Ultrasonography With Cardiovascular Risk Factors-A "SPIDER" Promoting Atherosclerosis. Front Cardiovasc Med 2021; 8:706490. [PMID: 34447790 PMCID: PMC8382941 DOI: 10.3389/fcvm.2021.706490] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/19/2021] [Indexed: 12/18/2022] Open
Abstract
Insufficient recommendations do not support the clinical use of carotid ultrasonography for further risk stratification in moderate-to-high risk patients with cardiovascular disease (CVD). A literature review was performed to assess six aspects of the research progress and limitations of carotid ultrasonography and carotid atherosclerosis-related risk factors: (1) structures of the carotid intima and media; (2) plaques; (3) inflammation; (4) dynamics of carotid blood flow; (5) early detection and intervention; and (6) risk factors for CVD. Although carotid intima-media thickness and carotid plaques are well-acknowledged independent predictors of CVD risk, normative and cut-off values are difficult to define due to the heterogeneous measurements reported in previous studies. Plaque properties, including location, number, density, and size, become more important risk predictors for cardiovascular disease, but a better approach for clinical use needs to be further established. Three-dimensional ultrasound and contrast-enhanced ultrasound are promising for promoting risk stratification with more details on plaque morphology. Moreover, inflammatory diseases and biomarkers should be evaluated for a full assessment of the inflammatory burden for atherosclerosis. Carotid flow velocity is not only an indicator for stenosis but also a potential risk predictor. Carotid atherosclerosis should be detected and treated early, and additional clinical trials are needed to determine the efficacy of these measures in reducing CVD risk. Cardiovascular risk factors tend to affect carotid plaques, and early treat-to-target therapy might yield clinical benefits. Based on the aforementioned six aspects, we consider that these six important factors act like a “SPIDER” spinning the web of atherosclerosis; a timely comprehensive assessment and intervention may halt the progression to CVD. Carotid ultrasound results should be combined with other atherosclerotic factors, and a comprehensive risk assessment may help to guide cardiovascular prevention decisions.
Collapse
Affiliation(s)
- Hongwei Li
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Xiaolin Xu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Baoming Luo
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuling Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| |
Collapse
|
11
|
Liao B, Geng L, Zhang F, Shu L, Wei L, Yeung PKK, Lam KSL, Chung SK, Chang J, Vanhoutte PM, Xu A, Wang K, Hoo RLC. Adipocyte fatty acid-binding protein exacerbates cerebral ischaemia injury by disrupting the blood-brain barrier. Eur Heart J 2021; 41:3169-3180. [PMID: 32350521 PMCID: PMC7556749 DOI: 10.1093/eurheartj/ehaa207] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/12/2019] [Accepted: 03/11/2020] [Indexed: 01/06/2023] Open
Abstract
Aims Adipocyte fatty acid-binding protein (A-FABP) is an adipokine implicating in various metabolic diseases. Elevated circulating levels of A-FABP correlate positively with poor prognosis in ischaemic stroke (IS) patients. No information is available concerning the role of A-FABP in the pathogenesis of IS. Experiments were designed to determine whether or not A-FABP mediates blood–brain barrier (BBB) disruption, and if so, to explore the molecular mechanisms underlying this deleterious effects. Methods and results Circulating A-FABP and its cerebral expression were increased in mice after middle cerebral artery occlusion. Genetic deletion and pharmacological inhibition of A-FABP alleviated cerebral ischaemia injury with reduced infarction volume, cerebral oedema, neurological deficits, and neuronal apoptosis; BBB disruption was attenuated and accompanied by reduced degradation of tight junction proteins and induction of matrix metalloproteinases-9 (MMP-9). In patients with acute IS, elevated circulating A-FABP levels positively correlated with those of MMP-9 and cerebral infarct volume. Mechanistically, ischaemia-induced elevation of A-FABP selectively in peripheral blood monocyte-derived macrophages and cerebral resident microglia promoted MMP-9 transactivation by potentiating JNK/c-Jun signalling, enhancing degradation of tight junction proteins and BBB leakage. The detrimental effects of A-FABP were prevented by pharmacological inhibition of MMP-9. Conclusion A-FABP is a key mediator of cerebral ischaemia injury promoting MMP-9-mediated BBB disruption. Inhibition of A-FABP is a potential strategy to improve IS outcome. ![]()
Collapse
Affiliation(s)
- Boya Liao
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Leiluo Geng
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Fang Zhang
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Guangzhou 510120, China
| | - Lingling Shu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ling Wei
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Anhui Province Key Laboratory on Cognition and Neuropsychiatric Disorders, No. 218, Jixi Road, Hefei, Anhui Province 230022, China
| | - Patrick K K Yeung
- Department of Anatomy, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Karen S L Lam
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Sookja K Chung
- Department of Anatomy, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Macau University of Science and Technology, Avenida Wai Long, Taipa 999078, Macau, China
| | - Junlei Chang
- Centre for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Blvd, Xili Nanshan, Shenzhen 518055, China
| | - Paul M Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kai Wang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Anhui Province Key Laboratory on Cognition and Neuropsychiatric Disorders, No. 218, Jixi Road, Hefei, Anhui Province 230022, China
| | - Ruby L C Hoo
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
12
|
Lee CH, Lui DTW, Lam KSL. Adipocyte Fatty Acid-Binding Protein, Cardiovascular Diseases and Mortality. Front Immunol 2021; 12:589206. [PMID: 33815359 PMCID: PMC8017191 DOI: 10.3389/fimmu.2021.589206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
It has been increasingly recognized that inflammation plays an important role in the pathogenesis of cardiovascular disease (CVD). In obesity, adipose tissue inflammation, especially in the visceral fat depots, contributes to systemic inflammation and promotes the development of atherosclerosis. Adipocyte fatty acid-binding protein (AFABP), a lipid chaperone abundantly secreted from the adipocytes and macrophages, is one of the key players mediating this adipose-vascular cross-talk, in part via its interaction with c-Jun NH2-terminal kinase (JNK) and activator protein-1 (AP-1) to form a positive feedback loop, and perpetuate inflammatory responses. In mice, selective JNK inactivation in the adipose tissue significantly reduced the expression of AFABP in their adipose tissue, as well as circulating AFABP levels. Importantly, fat transplant experiments showed that adipose-specific JNK inactivation in the visceral fat was sufficient to protect mice with apoE deficiency from atherosclerosis, with the beneficial effects attenuated by the continuous infusion of recombinant AFABP, supporting the role of AFABP as the link between visceral fat inflammation and atherosclerosis. In humans, raised circulating AFABP levels are associated with incident metabolic syndrome, type 2 diabetes and CVD, as well as non-alcoholic steatohepatitis, diabetic nephropathy and adverse renal outcomes, all being conditions closely related to inflammation and enhanced CV mortality. Collectively, these clinical data have provided support to AFABP as an important adipokine linking obesity, inflammation and CVD. This review will discuss recent findings on the role of AFABP in CVD and mortality, the possible underlying mechanisms, and pharmacological inhibition of AFABP as a potential strategy to combat CVD.
Collapse
Affiliation(s)
- Chi-Ho Lee
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, Hong Kong
| | - David T W Lui
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Karen S L Lam
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
13
|
Fosheim IK, Johnsen GM, Alnaes-Katjavivi P, Turowski G, Sugulle M, Staff AC. Decidua basalis and acute atherosis: Expression of atherosclerotic foam cell associated proteins. Placenta 2021; 107:1-7. [PMID: 33725567 DOI: 10.1016/j.placenta.2021.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Uteroplacental acute atherosis is frequently observed in preeclampsia, and shares features with early atherosclerotic lesions, including artery wall foam cells. The lipid-associated proteins FABP4 (fatty acid binding protein 4), perilipin-2, and LOX-1 (lectin-like oxidized LDL-receptor 1) are involved in atherosclerotic foam cell formation. Increased levels of these proteins have been associated with preeclampsia systemically and in placental tissue. Their role in acute atherosis is yet unidentified. Our aim was to describe the presence of these proteins in acute atherosis, and compare our findings to what is known in early atherosclerotic lesions. METHODS Serial sections of decidua basalis tissue from 12 normotensive (4 with acute atherosis) and 23 preeclamptic pregnancies (16 with acute atherosis) were stained with HE and immunostained for CK7, CD68, FABP4, perilipin-2, and LOX-1. Artery wall and perivascular protein expression was assessed in 190 spiral artery sections; 55 with acute atherosis. RESULTS Acute atherosis foam cells were commonly positive for perilipin-2 (55%), less often for FABP4 (13%), and never for LOX-1. LOX-1 was frequently observed in intramural trophoblasts of normal spiral arteries. Perivascularly, LOX-1 positivity of decidual stromal cells surrounding arteries with acute atherosis was significantly increased as compared to arteries lacking acute atherosis (38% vs. 15%, p < 0.001). DISCUSSION We found that perilipin-2 and FABP4 are expressed by acute atherosis foam cells, similar to atherosclerosis, supporting possible shared pathways for foam cell generation. Unlike atherosclerosis, LOX-1 is not present in acute atherosis, possibly explained by pregnancy-specific routes to decidua basalis foam cell generation.
Collapse
Affiliation(s)
- I K Fosheim
- Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - G M Johnsen
- Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway
| | - P Alnaes-Katjavivi
- Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway
| | - G Turowski
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - M Sugulle
- Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - A C Staff
- Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway.
| |
Collapse
|
14
|
Yang XL, Mi JH, Dong Q. FABP4 alleviates endoplasmic reticulum stress-mediated ischemia-reperfusion injury in PC12 cells via regulation of PPARγ. Exp Ther Med 2021; 21:181. [PMID: 33488790 PMCID: PMC7812580 DOI: 10.3892/etm.2021.9612] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/03/2020] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke is a life-threatening complication with a high rate of morbidity. Circulating fatty acid binding protein 4 (FABP4) has been reported to be associated with the outcome of acute ischemic stroke. The present study aimed to illustrate the function of FABP4 in ischemic stroke. PC12 cells exposed to oxygen glucose deprivation/reoxygenation (OGD/R) were used to mimic ischemia-reperfusion (I/R) injury in ischemic stroke. Cell viability was estimated using a Cell Counting Kit-8 assay. The expression of FABP4 in PC12 cells under OGD/R was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). PC12 cells were transfected with FABP4 small interfering RNA (siRNA), inflammatory cytokines and reactive oxygen species (ROS) were determined via RT-qPCR and ROS assay kit. Western blotting was performed to detect endoplasmic reticulum stress (ERS)-related proteins and peroxisome proliferator-activated receptor γ (PPARγ). Flow cytometry was used to evaluate the cell apoptotic rate. The expression of FABP4 increased gradually with the prolongation of reoxygenation within 8 h. FABP4-knockdown inhibited the transcription of inflammatory cytokines, the production of ROS and decreased cell apoptosis. Furthermore, decreased ERS-related proteins and increased PPARγ were estimated in PC12 cells transfected with FABP4 siRNA. PPARγ inhibitor GW9662 weakened the anti-apoptotic effect of FABP4-knockdown. Taken together, these results indicated that FABP4-knockdown suppressed cell apoptosis via relieving ERS; this effect was reversed by treatment of GW9662.
Collapse
Affiliation(s)
- Xiao-Lan Yang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201100, P.R. China
| | - Jian-Hua Mi
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201100, P.R. China
| | - Qing Dong
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201100, P.R. China
| |
Collapse
|
15
|
Hansen C, Sastre C, Wolcott Z, Bevers MB, Kimberly WT. Time-dependent, dynamic prediction of fatty acid-binding protein 4, Galectin-3, and soluble ST2 measurement with poor outcome after acute stroke. Int J Stroke 2020; 16:660-668. [PMID: 33167787 DOI: 10.1177/1747493020971166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Time-dependent change in the level of biomarkers after stroke is not well understood. We sought to compare fatty acid-binding protein 4 (FABP4), Galectin-3, and soluble ST2 to ascertain for a change in prediction of outcome at admission and 48 h later. METHODS Plasma FABP4, Galectin-3, and soluble ST2 were measured in biospecimens from acute stroke patients at the time of admission (n = 383) and 48 h later (n = 244). Functional outcome was assessed at 90 days using the modified Rankin Scale and dichotomized into good (modified Rankin Scale 0-2) and poor outcome (modified Rankin Scale 3-6). RESULTS On admission, elevated levels of each biomarker predicted poor outcome (FABP4: OR 1.92, 95% CI 1.42-2.59, P < 0.0001; Galectin-3: OR 1.85, 95% CI 1.42-2.40, P < 0.0001; soluble ST2: OR 1.55, 95% CI 1.22-1.97, P < 0.0001) and death (FABP4: OR 2.45; 95% CI 1.51-3.98; P < 0.0001; Galectin-3: OR 2.12; 95% CI 1.50-3.30; P < 0.0001; soluble ST2: OR 2.17; 95% CI 1.58-2.99; P < 0.0001). At 48 h, soluble ST2 predicted poor outcome (OR 2.62, 95% CI 1.77-3.88, P < 0.0001) and mortality (OR 3.36, 95% CI 2.06-5.48, P < 0.0001), and Galectin-3 predicted mortality only (OR 1.81, 95% CI 1.05-3.10, P = 0.033). FABP4 measured at 48 h was not predictive of outcome or death. Associations of Galectin-3 and soluble ST2 with outcome or mortality were independent of age, sex, and NIHSS, whereas those with FABP4 were not. CONCLUSIONS Galectin-3 performed better when measured on admission, whereas soluble ST2 was predictive at admission and better at 48 h after stroke. The time-dependent differences may reflect the evolving role of these pathways after acute stroke.
Collapse
Affiliation(s)
- Christina Hansen
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Cristina Sastre
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Zoe Wolcott
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew B Bevers
- Divisions of Stroke, Cerebrovascular and Critical Care Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - W Taylor Kimberly
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
16
|
Search for Reliable Circulating Biomarkers to Predict Carotid Plaque Vulnerability. Int J Mol Sci 2020; 21:ijms21218236. [PMID: 33153204 PMCID: PMC7662861 DOI: 10.3390/ijms21218236] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is responsible for 20% of ischemic strokes, and the plaques from the internal carotid artery the most frequently involved. Lipoproteins play a key role in carotid atherosclerosis since lipid accumulation contributes to plaque progression and chronic inflammation, both factors leading to plaque vulnerability. Carotid revascularization to prevent future vascular events is reasonable in some patients with high-grade carotid stenosis. However, the degree of stenosis alone is not sufficient to decide upon the best clinical management in some situations. In this context, it is essential to further characterize plaque vulnerability, according to specific characteristics (lipid-rich core, fibrous cap thinning, intraplaque hemorrhage). Although these features can be partly detected by imaging techniques, identifying carotid plaque vulnerability is still challenging. Therefore, the study of circulating biomarkers could provide adjunctive criteria to predict the risk of atherothrombotic stroke. In this regard, several molecules have been found altered, but reliable biomarkers have not been clearly established yet. The current review discusses the concept of vulnerable carotid plaque, and collects existing information about putative circulating biomarkers, being particularly focused on lipid-related and inflammatory molecules.
Collapse
|
17
|
Eslava-Alcon S, Extremera-García MJ, González-Rovira A, Rosal-Vela A, Rojas-Torres M, Beltran-Camacho L, Sanchez-Gomar I, Jiménez-Palomares M, Alonso-Piñero JA, Conejero R, Doiz E, Olarte J, Foncubierta-Fernández A, Lozano E, García-Cozar FJ, Rodríguez-Piñero M, Alvarez-Llamas G, Duran-Ruiz MC. Molecular signatures of atherosclerotic plaques: An up-dated panel of protein related markers. J Proteomics 2020; 221:103757. [PMID: 32247173 DOI: 10.1016/j.jprot.2020.103757] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/14/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis remains the leading cause of ischemic syndromes such as myocardial infarction or brain stroke, mainly promoted by plaque rupture and subsequent arterial blockade. Identification of vulnerable or high-risk plaques constitutes a major challenge, being necessary to identify patients at risk of occlusive events in order to provide them with appropriate therapies. Clinical imaging tools have allowed the identification of certain structural indicators of prone-rupture plaques, including a necrotic lipidic core, intimal and adventitial inflammation, extracellular matrix dysregulation, and smooth muscle cell depletion and micro-calcification. Additionally, alternative approaches focused on identifying molecular biomarkers of atherosclerosis have also been applied. Among them, proteomics has provided numerous protein markers currently investigated in clinical practice. In this regard, it is quite uncertain that a single molecule can describe plaque rupture, due to the complexity of the process itself. Therefore, it should be more accurate to consider a set of markers to define plaques at risk. Herein, we propose a selection of 76 proteins, from classical inflammatory to recently related markers, all of them identified in at least two proteomic studies analyzing unstable atherosclerotic plaques. Such panel could be used as a prognostic signature of plaque instability.
Collapse
Affiliation(s)
- S Eslava-Alcon
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain
| | - M J Extremera-García
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain
| | - A González-Rovira
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain
| | - A Rosal-Vela
- Institute of Biomedical Research Cadiz (INIBICA), Spain
| | - M Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain
| | - L Beltran-Camacho
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain
| | | | - M Jiménez-Palomares
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain
| | - J A Alonso-Piñero
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain
| | - R Conejero
- Angiology & Vascular Surgery Unit, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - E Doiz
- Angiology & Vascular Surgery Unit, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - J Olarte
- Angiology & Vascular Surgery Unit, Virgen Macarena Hospital, Seville, Spain
| | - A Foncubierta-Fernández
- Institute of Biomedical Research Cadiz (INIBICA), Spain; UGC Joaquín Pece, Distrito Sanitario Bahía de Cádiz-La Janda, Cádiz, Spain
| | - E Lozano
- Institute of Biomedical Research Cadiz (INIBICA), Spain; Internal Medicine Unit, Hospital de Jerez, Jerez, Spain
| | - F J García-Cozar
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain
| | - M Rodríguez-Piñero
- Angiology & Vascular Surgery Unit, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - G Alvarez-Llamas
- Immunology Department, IIS-Fundación Jimenez Diaz-UAM, Madrid, Spain; REDINREN, Madrid, Spain
| | - M C Duran-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Cadiz University, Spain; Institute of Biomedical Research Cadiz (INIBICA), Spain.
| |
Collapse
|
18
|
Luo YG, Han B, Sun TW, Liu X, Liu J, Zhang J. The association between serum adipocyte fatty acid-binding protein and 3-month disability outcome after aneurysmal subarachnoid hemorrhage. J Neuroinflammation 2020; 17:66. [PMID: 32075656 PMCID: PMC7029438 DOI: 10.1186/s12974-020-01743-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Adipocyte fatty acid-binding protein (FABP4) is an adipokine that plays an important role in development of cardiovascular and metabolic diseases. The aim of this study was to assess the 3-month prognostic value of serum levels of FABP4 in Chinese patients with aneurysmal subarachnoid hemorrhage (aSAH) on hospital admission. METHODS This was a prospective observational study from a stroke treatment center in Zhengzhou, China. From October 2016 to May 2018, patients with aSAH who were hospitalized within 24 h were included. In addition, 202 age- and gender-matched healthy volunteers were assigned to the healthy control group. At admission, serum levels of FABP4 were measured, and patients' characteristics, Hunt-Hess grade, and modified Fisher grade evaluated. At 3-month follow-up, functional outcome (Glasgow Outcome Scale score; dichotomized as poor [score 1-3] or good [score 4-5]) and all-cause mortality were recorded. Univariate and multivariate logistic regression models were used to investigate the association of FABP4 with the two endpoints. RESULTS A total of 418 patients with aSAH were included in this study. The median age was 58 years (interquartile range, 49-66 years), and 57.9% were women. FABP4 serum levels were related to Hunt-Hess score (r[Spearman] = 0.381; P < 0.001). Patients with a poor outcome and non-survivors had significantly increased serum FABP4 levels on admission (P < 0.001 for all). In multivariate logistic regression analysis, FABP4 was an independent predictor of poor outcome and mortality, with increased risks of 7% (odds ratios 1.07, 95% confidence interval [CI] 1.02-1.13; P = 0.001) and 5% (odds ratio 1.05, 95% CI, 1.01-1.12; P = 0.003), respectively. Receiver operating characteristics to predict functional outcome and mortality were significantly different between conventional risk factors (difference area under the curve 0.024, 95% CI 0.018-0.032) and FABP4 plus conventional risk factors (area under the curve 0.015, 95%CI 0.011-0.020). After FABP4 was added to the existing risk factors, mortality was better reclassified and was associated with the net reclassification improvement statistic (P = 0.009), while poor outcome was better reclassified and associated with both the integrated discrimination improvement and net reclassification improvement statistics (P < 0.05 for all). CONCLUSIONS Elevated serum FABP4 levels were related to poor outcome and mortality in a cohort of patients with aSAH.
Collapse
Affiliation(s)
- Yong-Gang Luo
- Department of Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Bing Han
- Department of Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Tong-Wen Sun
- Department of Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jun Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jun Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
19
|
Review of serum biomarkers in carotid atherosclerosis. J Vasc Surg 2019; 71:329-341. [PMID: 31327598 DOI: 10.1016/j.jvs.2019.04.488] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/23/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Carotid artery atherosclerotic stenosis is a preventable major cause of stroke, but there is still a need for definition of high-risk plaque in asymptomatic patients who might benefit from interventional therapies. Several image markers are recommended to characterize unstable plaques. The measurement of serum biomarkers is a promising method to assist in decision making, but the lack of robust evidence in the carotid environment burdens their potential as a standard of care. The goal of this review was to offer an updated state-of-the-art study of available serum biomarkers with clinical implications, with focus on those that may predict carotid symptom development. METHODS The Cochrane Library and MEDLINE databases were searched (all until September 2018) for studies on carotid plaque and serum biomarkers of atherosclerosis. Nonhuman, basic science, and histology studies were excluded, focusing on clinical studies. Selected abstracts were screened to include the most relevant articles on atherosclerotic plaque presence, progression, instability or symptom development. RESULTS Some well-established biomarkers for coronary disease are not relevant to carotid atherosclerosis and other inflammatory biomarkers, lipids, interleukins, homocysteine, and adipokines may be useful in quantifying carotid disease-related risk. Some serum biomarkers combined with image features may assist vascular specialists in selecting patients at high risk for stroke and in need of intervention. CONCLUSIONS Prospective studies applying a combination of biomarkers are essential to prove clinical usefulness.
Collapse
|
20
|
Bruen R, Curley S, Kajani S, Lynch G, O'Reilly ME, Dillon ET, Fitzsimons S, Mthunzi L, McGillicuddy FC, Belton O. Different monocyte phenotypes result in proresolving macrophages in conjugated linoleic acid-induced attenuated progression and regression of atherosclerosis. FASEB J 2019; 33:11006-11020. [PMID: 31284764 DOI: 10.1096/fj.201900922r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Monocytes/macrophages drive progression and regression of atherosclerosis. Conjugated linoleic acid (CLA), an anti-inflammatory lipid, mediates atheroprotective effects. We investigated how CLA alters monocyte/macrophage phenotype during attenuated progression and regression of atherosclerosis. Apolipoprotein E knockout (ApoE-/-) mice were fed a high-fat (60%) high-cholesterol (1%) diet (HFHCD) for 2 wk, followed by 6-wk 1% CLA 80:20 supplementation to investigate disease progression. Simultaneously, ApoE-/- mice were fed a 12-wk HFHCD with/without CLA for the final 4 wk to investigate regression. Aortic lesions were quantified by en face staining. Proteomic analysis, real-time quantitative PCR and flow cytometry were used to interrogate monocyte/macrophage phenotypes. CLA supplementation inhibited atherosclerosis progression coincident with decreased proinflammatory and increased anti-inflammatory macrophages. However, CLA-induced regression was associated with increased proinflammatory monocytes resulting in increased proresolving M2 bone marrow-derived macrophages, splenic macrophages, and dendritic cells in lesion-draining lymph nodes. Proteomic analysis confirmed regulation of a proinflammatory bone marrow response, which was abolished upon macrophage differentiation. Thus, in attenuation and regression of atherosclerosis, regardless of the monocyte signature, during monocyte to macrophage differentiation, proresolving macrophages prevail, mediating vascular repair. This study provides novel mechanistic insight into the monocyte/macrophage phenotypes in halted atherosclerosis progression and regression of atherosclerosis.-Bruen, R., Curley, S., Kajani, S., Lynch, G., O'Reilly, M. E., Dillon, E. T., Fitzsimons, S., Mthunzi, L., McGillicuddy, F. C., Belton, O. Different monocyte phenotypes result in proresolving macrophages in conjugated linoleic acid-induced attenuated progression and regression of atherosclerosis.
Collapse
Affiliation(s)
- Robyn Bruen
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Seán Curley
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Sarina Kajani
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Gina Lynch
- School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Dublin, Ireland
| | - Marcella E O'Reilly
- School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Dublin, Ireland
| | - Eugéne T Dillon
- Mass Spectrometry Resource, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Stephen Fitzsimons
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Liberty Mthunzi
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Fiona C McGillicuddy
- Diabetes Complications Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Orina Belton
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Li B, Wu J, Jiang P, Li M, Liu Q, Cao Y, Wang S. Serum fatty acid binding protein 4 is positively associated with early stroke recurrence in nondiabetic ischemic stroke. Aging (Albany NY) 2019; 11:1977-1989. [PMID: 30969942 PMCID: PMC6503889 DOI: 10.18632/aging.101886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/20/2019] [Indexed: 12/14/2022]
Abstract
Adipocyte fatty acid–binding protein (FABP4) played critical roles in metabolic syndrome, inflammatory responses and cardiovascular diseases. It aimed to investigate the associations of serum FABP4 levels with early stroke recurrence. This study included the 206 acute ischemic stroke patients hospitalized in our institution. Stroke recurrence events were assessed at the 3-month follow-up. The median of FABP level was 22.6 (IQR, 17.9-31.6) ng/mL in patients with stroke recurrence (N=36), which was higher than in patients without stroke recurrence [16.9 (IQR, 11.8-21.4) ng/mL] (P<0.001). As a continuous variable, the unadjusted and adjusted risk of stroke recurrence would be increased by 12% (OR=1.12 [95% CI 1.06–1.17], P<0.001) and 8% (1.08 [1.02–1.14], P=0.006) for every 1 ng/ml increment of FABP4. The Area under the curve (AUC) of serum FABP4 and NIH Stroke Scale (NIHSS) score for predicting stroke recurrence was 0.73 (95% CI: 0.64–0.82) and 0.72 (95% CI: 0.64–0.81), presenting no discriminating capacity (P=0.45). In the combining model, the AUC of NIHSS score was further improved to 0.77 by FABP4 (0.77; 95% CI: 0.69–0.85), which was significant (P=0.01). The risk of stroke recurrence can be predicted by elevated FABP4 levels in serum of nondiabetic patients with first-ever ischemic stroke.
Collapse
Affiliation(s)
- Bo Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Jun Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Pengjun Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Maogui Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Qingyuan Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Yong Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| |
Collapse
|
22
|
Trojnar M, Patro-Małysza J, Kimber-Trojnar Ż, Leszczyńska-Gorzelak B, Mosiewicz J. Associations between Fatty Acid-Binding Protein 4⁻A Proinflammatory Adipokine and Insulin Resistance, Gestational and Type 2 Diabetes Mellitus. Cells 2019; 8:cells8030227. [PMID: 30857223 PMCID: PMC6468522 DOI: 10.3390/cells8030227] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/03/2019] [Accepted: 03/03/2019] [Indexed: 12/12/2022] Open
Abstract
There is ample scientific evidence to suggest a link between the fatty acid-binding protein 4 (FABP4) and insulin resistance, gestational (GDM), and type 2 (T2DM) diabetes mellitus. This novel proinflammatory adipokine is engaged in the regulation of lipid metabolism at the cellular level. The molecule takes part in lipid oxidation, the regulation of transcription as well as the synthesis of membranes. An involvement of FABP4 in the pathogenesis of obesity and insulin resistance seems to be mediated via FABP4-dependent peroxisome proliferator-activated receptor γ (PPARγ) inhibition. A considerable number of studies have shown that plasma concentrations of FABP4 is increased in obesity and T2DM, and that circulating FABP4 levels are correlated with certain clinical parameters, such as body mass index, insulin resistance, and dyslipidemia. Since plasma-circulating FABP4 has the potential to modulate the function of several types of cells, it appears to be of extreme interest to try to develop potential therapeutic strategies targeting the pathogenesis of metabolic diseases in this respect. In this manuscript, representing a detailed review of the literature on FABP4 and the abovementioned metabolic disorders, various mechanisms of the interaction of FABP4 with insulin signaling pathways are thoroughly discussed. Clinical aspects of insulin resistance in diabetic patients, including women diagnosed with GDM, are analyzed as well.
Collapse
Affiliation(s)
- Marcin Trojnar
- Chair and Department of Internal Medicine, Medical University of Lublin, 20-081 Lublin, Poland.
| | - Jolanta Patro-Małysza
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland.
| | - Żaneta Kimber-Trojnar
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland.
| | | | - Jerzy Mosiewicz
- Chair and Department of Internal Medicine, Medical University of Lublin, 20-081 Lublin, Poland.
| |
Collapse
|
23
|
Nunes HC, Scarano WR, Deffune E, Felisbino SL, Porreca I, Delella FK. Bisphenol a and mesenchymal stem cells: Recent insights. Life Sci 2018; 206:22-28. [DOI: 10.1016/j.lfs.2018.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 02/08/2023]
|
24
|
Gharpure KM, Pradeep S, Sans M, Rupaimoole R, Ivan C, Wu SY, Bayraktar E, Nagaraja AS, Mangala LS, Zhang X, Haemmerle M, Hu W, Rodriguez-Aguayo C, McGuire M, Mak CSL, Chen X, Tran MA, Villar-Prados A, Pena GA, Kondetimmanahalli R, Nini R, Koppula P, Ram P, Liu J, Lopez-Berestein G, Baggerly K, S Eberlin L, Sood AK. FABP4 as a key determinant of metastatic potential of ovarian cancer. Nat Commun 2018; 9:2923. [PMID: 30050129 PMCID: PMC6062524 DOI: 10.1038/s41467-018-04987-y] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/06/2018] [Indexed: 12/30/2022] Open
Abstract
The standard treatment for high-grade serous ovarian cancer is primary debulking surgery followed by chemotherapy. The extent of metastasis and invasive potential of lesions can influence the outcome of these primary surgeries. Here, we explored the underlying mechanisms that could increase metastatic potential in ovarian cancer. We discovered that FABP4 (fatty acid binding protein) can substantially increase the metastatic potential of cancer cells. We also found that miR-409-3p regulates FABP4 in ovarian cancer cells and that hypoxia decreases miR-409-3p levels. Treatment with DOPC nanoliposomes containing either miR-409-3p mimic or FABP4 siRNA inhibited tumor progression in mouse models. With RPPA and metabolite arrays, we found that FABP4 regulates pathways associated with metastasis and affects metabolic pathways in ovarian cancer cells. Collectively, these findings demonstrate that FABP4 is functionally responsible for aggressive patterns of disease that likely contribute to poor prognosis in ovarian cancer.
Collapse
Affiliation(s)
- Kshipra M Gharpure
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Marta Sans
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Rajesha Rupaimoole
- Department of Pathology and Institute of RNA Medicine, Beth Israel Deaconess Medical Center Cancer Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Cristina Ivan
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, 77030, Texas, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sherry Y Wu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Emine Bayraktar
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Archana S Nagaraja
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, 77030, Texas, USA
| | - Xinna Zhang
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, 77030, Texas, USA
| | - Monika Haemmerle
- Martin-Luther-University Halle-Wittenberg, Institute of Pathology, 06112, Halle (Saale), Germany
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Cristian Rodriguez-Aguayo
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, 77030, Texas, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael McGuire
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Celia Sze Ling Mak
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Xiuhui Chen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Michelle A Tran
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Alejandro Villar-Prados
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Guillermo Armaiz Pena
- Department of Pharmacology, Ponce Health Sciences University, Ponce, 00716, Puerto Rico
| | | | - Ryan Nini
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Pranavi Koppula
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Prahlad Ram
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jinsong Liu
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gabriel Lopez-Berestein
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, 77030, Texas, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Keith Baggerly
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Livia S Eberlin
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA.
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, 77030, Texas, USA.
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Li S, Bi P, Zhao W, Lian Y, Zhu H, Xu D, Ding J, Wang Q, Yin C. Prognostic Utility of Fatty Acid-Binding Protein 4 in Patients with Type 2 Diabetes and Acute Ischemic Stroke. Neurotox Res 2017; 33:309-315. [PMID: 28801883 DOI: 10.1007/s12640-017-9792-z] [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: 06/06/2017] [Revised: 07/13/2017] [Accepted: 07/28/2017] [Indexed: 12/11/2022]
Abstract
The role of fatty acid-binding proteins (FABPs) in atherosclerosis has been investigated. The aim of this study was to verify the hypothesis that higher levels of serum fatty acid-binding protein 4 (FABP4) could be a prognostic factor in Chinese patients with type 2 diabetes (T2DM) and acute ischemic stroke (AIS). From September 2015 to August 2016, consecutive first-ever AIS patients combined with T2DM were included in this study. FABP4, NIH stroke scale (NIHSS), and conventional risk factors were evaluated to determine their value to predict functional outcomes within 3 months. Multivariate analyses were performed using logistic regression models. We measured FABP4 in 329 patients. The median age of patients included in this study was 63 (IQR, 56-72) years and 45.9% were women. FABP4 serum levels were obtained at a median of 8.5 h (IQR, 4.0-14.0 h) after the stroke onset with a median value of 21.4 ng/ml (IQR, 15.6-28.2 ng/ml). In multivariable models, FABP4 remained an independent stroke severity predictor with an adjusted OR of 1.05 (95% CI, 1.02-1.09). In multivariate models comparing the third (odd ratio (OR), 2.25; 95% confidence interval (CI), 1.59-3.54) and fourth quartiles (OR, 3.75; 95% CI, 2.48-5.03) against the first quartile of the FABP4, levels of FABP4 were associated with poor functional outcome. At 3 months, 38 patients (11.6%; 95%CI, 8.1-15.0%) had died. The mortality distribution across the FABP4 quartiles ranged between 3.7% (first quartile) and 20.7% (fourth quartile). Elevation of FABP4 is associated with an increased risk of death and poor functional outcome events in patients with type 2 diabetes and acute ischemic stroke and is independent of other established clinical risk predictors and biomarkers.
Collapse
Affiliation(s)
- Siou Li
- Department of Endocrinology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Pengxiang Bi
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, No 5, Tongxiang Road, Aimin District, Mudanjiang, 157011, People's Republic of China
| | - Weina Zhao
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, No 5, Tongxiang Road, Aimin District, Mudanjiang, 157011, People's Republic of China
| | - Yifei Lian
- Department of Endocrinology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Haifu Zhu
- Department of Endocrinology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Dan Xu
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, No 5, Tongxiang Road, Aimin District, Mudanjiang, 157011, People's Republic of China
| | - Jiayuan Ding
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, No 5, Tongxiang Road, Aimin District, Mudanjiang, 157011, People's Republic of China
| | - Quankui Wang
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, No 5, Tongxiang Road, Aimin District, Mudanjiang, 157011, People's Republic of China
| | - Changhao Yin
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, No 5, Tongxiang Road, Aimin District, Mudanjiang, 157011, People's Republic of China.
| |
Collapse
|
26
|
Chang L, Zhang J, Liu L, Huang Z, Han Y, Zhu Y. Fatty acid binding protein 4 is associated with stroke risk and severity in patients with acute ischemic stroke. J Neuroimmunol 2017; 311:29-34. [PMID: 28781090 DOI: 10.1016/j.jneuroim.2017.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 07/13/2017] [Accepted: 07/18/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The role of fatty acid-binding proteins (FABPs) in atherosclerosis has been investigated. The aim of this study was to verify the hypothesis that higher levels of serum FABP4 could be a biomarker for stroke and associated with stroke severity in Chinese patients with ischemic stroke. METHODS All consecutive patients with first-ever acute ischemic stroke from September 2015 to August 2016 were recruited to participate in the study. Serum FABP4 levels and routine tests were examined at admission. The National Institutes of Health Stroke Scale (NIHSS) score was assessed on admission blinded to FABP4 levels. RESULTS We recorded 277 stroke patients. There was a significant difference in median serum FABP4 levels between stroke patients and control cases (P<0.001). Serum FABP4 levels increased with increasing severity of stroke as defined by the NIHSS score. The result illustrates the time course of serum FABP4, showing significant changes with day of sampling, with peak levels on day 1, falling to a plateau by days 2 to 5. At admission, 129 patients (46.6%) had a minor stroke (NIHSS≤5). In these patients, the median serum FABP4 level was lower than that observed in patients with moderate-to-sever clinical severity (P<0.001). In multivariate models comparing the second, third, and fourth quartiles against the first quartile of the FABP4, levels of FABP4 were associated with stroke risk and severity. CONCLUSION High levels of FABP4 are significantly related to stroke risk and severity, independent from other traditional and emerging risk factors, suggesting that they may play a role in stroke pathogenesis.
Collapse
Affiliation(s)
- Li Chang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
| | - Jianlong Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Li Liu
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhen Huang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yingbo Han
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yanyan Zhu
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
27
|
Liang C, Wang QS, Yang X, Niu N, Hu QQ, Zhang BL, Wu MM, Yu CJ, Chen X, Song BL, Zhang ZR, Ma HP. Oxidized low-density lipoprotein stimulates epithelial sodium channels in endothelial cells of mouse thoracic aorta. Br J Pharmacol 2017; 175:1318-1328. [PMID: 28480509 DOI: 10.1111/bph.13853] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/14/2017] [Accepted: 05/03/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The epithelial sodium channel (ENaC) is expressed in endothelial cells and acts as a negative modulator of vasodilatation. Oxidized LDL (ox-LDL) is a key pathological factor in endothelial dysfunction. In the present study we examined the role of ENaC in ox-LDL-induced endothelial dysfunction and its associated signal transduction pathway. EXPERIMENTAL APPROACH Patch clamp techniques combined with pharmacological approaches were used to examine ENaC activity in the endothelial cells of a split-open mouse thoracic aorta. Western blot analysis was used to determine ENaC expression in the aorta. The aorta relaxation was measured using a wire myograph assay. KEY RESULTS Ox-LDL, but not LDL, significantly increased ENaC activity in the endothelial cells attached to split-open thoracic aortas, and the increase was inhibited by a lectin-like ox-LDL receptor-1 (LOX-1) antagonist (κ-carrageenan), an NADPH oxidase inhibitor (apocynin), and a scavenger of ROS (TEMPOL). Sodium nitroprusside, an NO donor, diminished the ox-LDL-mediated activation of ENaC, and this effect was abolished by inhibiting soluble guanylate cyclase (sGC) and PKG. Ox-LDL reduced the endothelium-dependent vasodilatation of the aorta pectoralis induced by ACh, and this reduction was partially restored by blocking ENaC. CONCLUSION AND IMPLICATIONS Ox-LDL stimulates ENaC in endothelial cells through LOX-1 receptor-mediated activation of NADPH oxidase and accumulation of intracellular ROS. Since the stimulation of ENaC can be reversed by elevating NO, we suggest that both inhibition of ENaC and an elevation of NO may protect the endothelium from ox-LDL-induced dysfunction. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Chen Liang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Qiu-Shi Wang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Xu Yang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Na Niu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Qing-Qing Hu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Bao-Long Zhang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Ming-Ming Wu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Chang-Jiang Yu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Xiao Chen
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Bin-Lin Song
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Zhi-Ren Zhang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - He-Ping Ma
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
28
|
Rodríguez-Calvo R, Girona J, Alegret JM, Bosquet A, Ibarretxe D, Masana L. Role of the fatty acid-binding protein 4 in heart failure and cardiovascular disease. J Endocrinol 2017; 233:R173-R184. [PMID: 28420707 DOI: 10.1530/joe-17-0031] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/18/2017] [Indexed: 01/05/2023]
Abstract
Obesity and ectopic fat accumulation in non-adipose tissues are major contributors to heart failure (HF) and cardiovascular disease (CVD). Adipocytes act as endocrine organs by releasing a large number of bioactive molecules into the bloodstream, which participate in a communication network between white adipose tissue and other organs, including the heart. Among these molecules, fatty acid-binding protein 4 (FABP4) has recently been shown to increase cardiometabolic risk. Both clinical and experimental evidence have identified FABP4 as a relevant player in atherosclerosis and coronary artery disease, and it has been directly related to cardiac alterations such as left ventricular hypertrophy (LVH) and both systolic and diastolic cardiac dysfunction. The available interventional studies preclude the establishment of a direct causal role of this molecule in CVD and HF and propose FABP4 as a biomarker rather than as an aetiological factor. However, several experimental reports have suggested that FABP4 may act as a direct contributor to cardiac metabolism and physiopathology, and the pharmacological targeting of FABP4 may restore some of the metabolic alterations that are conducive to CVD and HF. Here, we review the current knowledge regarding FABP4 in the context of HF and CVD as well as the molecular basis by which this protein participates in the regulation of cardiac function.
Collapse
Affiliation(s)
- Ricardo Rodríguez-Calvo
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Josefa Girona
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Josep M Alegret
- Department of CardiologyCardiovascular Research Group, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Reus, Spain
| | - Alba Bosquet
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Daiana Ibarretxe
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Lluís Masana
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| |
Collapse
|
29
|
Flores AE, Pascotini ET, Kegler A, Gabbi P, Bochi GV, Barbisan F, Duarte T, Prado ALC, Duarte MMMF, da Cruz IBM, Moresco RN, Santos ARS, Bresciani G, Royes LFF, Fighera MR. ALA16VAL-MnSOD gene polymorphism and stroke: Association with dyslipidemia and glucose levels. Gene 2017; 627:57-62. [PMID: 28552711 DOI: 10.1016/j.gene.2017.05.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/25/2017] [Accepted: 05/24/2017] [Indexed: 01/25/2023]
Abstract
Stroke risk has been associated to the progression of carotid plaques due to high glucose levels and lipid accumulation, which are greatly associated to cerebral injury, brain oxidative stress, and apoptosis. The ALA16VAL-MnSOD gene single nucleotide polymorphism (SNP) has shown to modulate risk factors of several metabolic and vascular diseases, such as blood glucose (GLU) and lipid levels. However, the association of these factors in stroke patients has not been studied to date. Thus, we evaluated the influence of the Ala16Val-MnSOD SNP on lipid profile, GLU levels, oxidative and DNA damage of 44 patients in a late phase of stroke (>6months). The statistical analysis showed a greater proportion of VV carries in stroke patients. The results also indicated that stroke patients had higher cholesterol (CHO) and GLU levels when compared to healthy counterparts. Interestingly, V allele carriers with stroke showed higher levels of CHO and GLU when compared to AA stroke and healthy counterparts. Our findings suggest that oxidative stress markers are still increased even after 6 months of cerebral injury. Furthermore, we propose that the Ala16Val-MnSOD SNPs may contribute to hypercholesterolemia and higher GLU levels, increasing the risk to neurovascular events that may lead to stroke.
Collapse
Affiliation(s)
- Ariane Ethur Flores
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Eduardo Tanuri Pascotini
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Aline Kegler
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Patricia Gabbi
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Guilherme Vargas Bochi
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Fernanda Barbisan
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Thiago Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | | | - Marta M M F Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Ivana B M da Cruz
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Rafael Noal Moresco
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Adair Roberto Soares Santos
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Guilherme Bresciani
- Grupo de Investigación en Rendimiento Físico y Salud (IRyS), Escuela de Educación Física, Pontificia Universidad Católica de Valparaiso, Valparaiso, Chile
| | - Luiz Fernando Freire Royes
- Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil.
| |
Collapse
|
30
|
Tu WJ, Zeng XW, Deng A, Zhao SJ, Luo DZ, Ma GZ, Wang H, Liu Q. Circulating FABP4 (Fatty Acid-Binding Protein 4) Is a Novel Prognostic Biomarker in Patients With Acute Ischemic Stroke. Stroke 2017; 48:1531-1538. [PMID: 28487339 DOI: 10.1161/strokeaha.117.017128] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/04/2017] [Accepted: 04/10/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND PURPOSE FABP4 (fatty acid-binding protein 4) is an intracellular lipid chaperone involved in coordination of lipid transportation and atherogenesis. This study aimed at observing the effect of FABP4 on the 3-month outcomes in Chinese patients with acute ischemic stroke. METHODS In a prospective multicenter observational study, serum concentrations of FABP4 were on admission measured in plasma of 737 consecutive patients with acute ischemic stroke. Serum concentrations of FABP4, National Institutes of Health Stroke Scale score, and conventional risk factors were evaluated to determine their value to predict functional outcome and mortality within 3 months. RESULTS During follow-up, an unfavorable functional outcome was found in 260 patients (35.3%), and 94 patients (12.8%) died. In multivariate models comparing the third and fourth quartiles to the first quartile of FABP4, the concentrations of FABP4 were associated with poor functional outcome and mortality. Compared with the reference category (Q1-Q3), the concentrations of FABP4 in Q4 had a relative risk of 4.77 (95% confidence interval [CI], 2.02-8.15; P<0.001) for poor functional outcome and mortality (odds ratio, 6.15; 95% CI, 3.43-12.68) after adjusting for other significant outcome predictors in univariate logistic regression analysis. Receiver-operating characteristic curves to predict poor functional outcome and mortality demonstrated areas under the curve of FABP4 of 0.78 (95% CI, 0.75-0.82) and 0.83 (95% CI, 0.79-0.88), which improved the prognostic accuracy of National Institutes of Health Stroke Scale score with combined areas under the curve of 0.83 (95% CI, 0.76-0.89; P<0.01) and 0.86 (95% CI, 0.81-0.92), respectively. CONCLUSIONS Data show that FABP4 is a novel independent prognostic marker improving the currently used risk stratification of stroke patients.
Collapse
Affiliation(s)
- Wen-Jun Tu
- From the Institute of Radiation Medicine, China Academy of Medical Science and Peking Union Medical College, Tianjin, China (W.-J.T., Q.L.); Department of Neurology, China Rehabilitation Research Center, Beijing, China (W.-J.T., S.-J.Z.); School of Rehabilitation Medicine, Capital Medical University, Beijing, China (W.-J.T., H.W.); Department of Neurosurgery, Affiliated Hospital of Weifang Medical University, China (X.-W.Z., A.D.); and Department of Neurology, Provincial Hospital of Shandong University, Jinan, China (D.-Z.L., G.-Z.M.)
| | - Xian-Wei Zeng
- From the Institute of Radiation Medicine, China Academy of Medical Science and Peking Union Medical College, Tianjin, China (W.-J.T., Q.L.); Department of Neurology, China Rehabilitation Research Center, Beijing, China (W.-J.T., S.-J.Z.); School of Rehabilitation Medicine, Capital Medical University, Beijing, China (W.-J.T., H.W.); Department of Neurosurgery, Affiliated Hospital of Weifang Medical University, China (X.-W.Z., A.D.); and Department of Neurology, Provincial Hospital of Shandong University, Jinan, China (D.-Z.L., G.-Z.M.)
| | - Aijun Deng
- From the Institute of Radiation Medicine, China Academy of Medical Science and Peking Union Medical College, Tianjin, China (W.-J.T., Q.L.); Department of Neurology, China Rehabilitation Research Center, Beijing, China (W.-J.T., S.-J.Z.); School of Rehabilitation Medicine, Capital Medical University, Beijing, China (W.-J.T., H.W.); Department of Neurosurgery, Affiliated Hospital of Weifang Medical University, China (X.-W.Z., A.D.); and Department of Neurology, Provincial Hospital of Shandong University, Jinan, China (D.-Z.L., G.-Z.M.)
| | - Sheng-Jie Zhao
- From the Institute of Radiation Medicine, China Academy of Medical Science and Peking Union Medical College, Tianjin, China (W.-J.T., Q.L.); Department of Neurology, China Rehabilitation Research Center, Beijing, China (W.-J.T., S.-J.Z.); School of Rehabilitation Medicine, Capital Medical University, Beijing, China (W.-J.T., H.W.); Department of Neurosurgery, Affiliated Hospital of Weifang Medical University, China (X.-W.Z., A.D.); and Department of Neurology, Provincial Hospital of Shandong University, Jinan, China (D.-Z.L., G.-Z.M.)
| | - Ding-Zhen Luo
- From the Institute of Radiation Medicine, China Academy of Medical Science and Peking Union Medical College, Tianjin, China (W.-J.T., Q.L.); Department of Neurology, China Rehabilitation Research Center, Beijing, China (W.-J.T., S.-J.Z.); School of Rehabilitation Medicine, Capital Medical University, Beijing, China (W.-J.T., H.W.); Department of Neurosurgery, Affiliated Hospital of Weifang Medical University, China (X.-W.Z., A.D.); and Department of Neurology, Provincial Hospital of Shandong University, Jinan, China (D.-Z.L., G.-Z.M.)
| | - Guo-Zhao Ma
- From the Institute of Radiation Medicine, China Academy of Medical Science and Peking Union Medical College, Tianjin, China (W.-J.T., Q.L.); Department of Neurology, China Rehabilitation Research Center, Beijing, China (W.-J.T., S.-J.Z.); School of Rehabilitation Medicine, Capital Medical University, Beijing, China (W.-J.T., H.W.); Department of Neurosurgery, Affiliated Hospital of Weifang Medical University, China (X.-W.Z., A.D.); and Department of Neurology, Provincial Hospital of Shandong University, Jinan, China (D.-Z.L., G.-Z.M.)
| | - Hong Wang
- From the Institute of Radiation Medicine, China Academy of Medical Science and Peking Union Medical College, Tianjin, China (W.-J.T., Q.L.); Department of Neurology, China Rehabilitation Research Center, Beijing, China (W.-J.T., S.-J.Z.); School of Rehabilitation Medicine, Capital Medical University, Beijing, China (W.-J.T., H.W.); Department of Neurosurgery, Affiliated Hospital of Weifang Medical University, China (X.-W.Z., A.D.); and Department of Neurology, Provincial Hospital of Shandong University, Jinan, China (D.-Z.L., G.-Z.M.)
| | - Qiang Liu
- From the Institute of Radiation Medicine, China Academy of Medical Science and Peking Union Medical College, Tianjin, China (W.-J.T., Q.L.); Department of Neurology, China Rehabilitation Research Center, Beijing, China (W.-J.T., S.-J.Z.); School of Rehabilitation Medicine, Capital Medical University, Beijing, China (W.-J.T., H.W.); Department of Neurosurgery, Affiliated Hospital of Weifang Medical University, China (X.-W.Z., A.D.); and Department of Neurology, Provincial Hospital of Shandong University, Jinan, China (D.-Z.L., G.-Z.M.).
| |
Collapse
|
31
|
Pascotini ET, Flores AE, Kegler A, Gabbi P, Bochi GV, Algarve TD, Prado ALC, Duarte MM, da Cruz IB, Moresco RN, Royes LFF, Fighera MR. Apoptotic markers and DNA damage are related to late phase of stroke: Involvement of dyslipidemia and inflammation. Physiol Behav 2015; 151:369-78. [DOI: 10.1016/j.physbeh.2015.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/30/2015] [Accepted: 08/01/2015] [Indexed: 12/01/2022]
|
32
|
Markers of inflammation associated with plaque progression and instability in patients with carotid atherosclerosis. Mediators Inflamm 2015; 2015:718329. [PMID: 25960621 PMCID: PMC4415469 DOI: 10.1155/2015/718329] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 03/22/2015] [Indexed: 11/22/2022] Open
Abstract
Atherosclerosis is the focal expression of a systemic disease affecting medium- and large-sized arteries, in which traditional cardiovascular risk factor and immune factors play a key role. It is well accepted that circulating biomarkers, including C-reactive protein and interleukin-6, reliably predict major cardiovascular events, including myocardial infarction or death. However, the relevance of biomarkers of systemic inflammation to atherosclerosis progression in the carotid artery is less established. The large majority of clinical studies focused on the association between biomarkers and subclinical atherosclerosis, that is, carotid intima-media thickening (cIMT), which represents an earlier stage of the disease. The aim of this work is to review inflammatory biomarkers that were associated with a higher atherosclerotic burden, a faster disease progression, and features of plaque instability, such as inflammation or neovascularization, in patients with carotid atherosclerotic plaque, which represents an advanced stage of disease compared with cIMT. The association of biomarkers with the occurrence of cerebrovascular events, secondary to carotid plaque rupture, will also be presented. Currently, the degree of carotid artery stenosis is used to predict the risk of future cerebrovascular events in patients affected by carotid atherosclerosis. However, this strategy appears suboptimal. The identification of suitable biomarkers could provide a useful adjunctive criterion to ensure better risk stratification and optimize management.
Collapse
|
33
|
Boss M, Kemmerer M, Brüne B, Namgaladze D. FABP4 inhibition suppresses PPARγ activity and VLDL-induced foam cell formation in IL-4-polarized human macrophages. Atherosclerosis 2015; 240:424-30. [PMID: 25897794 DOI: 10.1016/j.atherosclerosis.2015.03.042] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/26/2015] [Accepted: 03/30/2015] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Macrophages, converted to lipid-loaded foam cells, accumulate in atherosclerotic lesions. Macrophage lipid metabolism is transcriptionally regulated by peroxisome proliferator-activated receptor gamma (PPARγ), and its target gene fatty acid binding protein 4 (FABP4) accelerates the progression of atherosclerosis in mouse models. Since expression of PPARγ and FABP4 is increased upon interleukin-4 (IL-4)-induced macrophage polarization, we aimed to investigate the role of FABP4 in human IL-4-polarized macrophages. METHODS AND RESULTS We investigated the impact of FABP4 on PPARγ-dependent gene expression in primary human monocytes differentiated to macrophages in the presence of IL-4. IL-4 increased PPARγ and its target genes lipoprotein lipase (LPL) and FABP4 compared to non-polarized or LPS/interferon γ-stimulated macrophages. LPL expression correlated with increased very low density lipoprotein (VLDL)-induced triglyceride accumulation in IL-4-polarized macrophages, which was sensitive to inhibition of lipolysis or PPARγ antagonism. Inhibition of FABP4 during differentiation using chemical inhibitors BMS309403 and HTS01037 or FABP4 siRNA decreased the expression of FABP4 and LPL, and reduced lipid accumulation in macrophages treated with VLDL. FABP4 or LPL inhibition also reduced the expression of inflammatory mediators chemokine (C-C motif) ligand 2 (CCL2) and IL-1β in response to VLDL in IL-4-polarized macrophages. PPARγ luciferase reporter assays confirmed that FABP4 supports fatty acid-induced PPARγ activation. CONCLUSION Our findings suggest that IL-4 induces a lipid-accumulating macrophage phenotype by activating PPARγ and subsequent LPL expression. Inhibition of FABP4 decreases VLDL-induced foam cell formation, indicating that anti-atherosclerotic effects achieved by FABP4 inhibition in mouse models may be feasible in the human system as well.
Collapse
Affiliation(s)
- Marcel Boss
- Institute of Biochemistry 1, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Marina Kemmerer
- Institute of Biochemistry 1, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Bernhard Brüne
- Institute of Biochemistry 1, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Dmitry Namgaladze
- Institute of Biochemistry 1, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
34
|
Miedema MD, Maziarz M, Biggs ML, Zieman SJ, Kizer JR, Ix JH, Mozaffarian D, Tracy RP, Psaty BM, Siscovick DS, Mukamal KJ, Djousse L. Plasma-free fatty acids, fatty acid-binding protein 4, and mortality in older adults (from the Cardiovascular Health Study). Am J Cardiol 2014; 114:843-8. [PMID: 25073566 PMCID: PMC4162821 DOI: 10.1016/j.amjcard.2014.06.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/14/2014] [Accepted: 06/14/2014] [Indexed: 02/07/2023]
Abstract
Plasma-free fatty acids (FFAs) are largely derived from adipose tissue. Elevated levels of FFA and fatty acid-binding protein 4 (FABP4), a key cytoplasmic chaperone of fatty acids, have been associated with adverse cardiovascular outcomes, but limited data are available on the relation of these biomarkers with cardiovascular and total mortality. We studied 4,707 participants with a mean age of 75 years who had plasma FFA and FABP4 measured in 1992 to 1993 as part of the Cardiovascular Health Study, an observational cohort of community-dwelling older adults. Over a median follow-up of 11.8 years, 3,555 participants died. Cox proportional hazard regression was used to determine the association between FFA, FABP4, and mortality. In fully adjusted models, FFA were associated with dose-dependent significantly higher total mortality (hazard ratio [HR] per SD: 1.14, 95% confidence interval [CI] 1.09 to 1.18), but FABP4 levels were not (HR 1.04, 95% CI 0.98 to 1.09). In a cause-specific mortality analysis, higher concentrations of FFA were associated with significantly higher risk of death because of cardiovascular disease, dementia, infection, and respiratory causes but not cancer or trauma. We did not find evidence of an interaction between FFA and FABP4 (p = 0.45), but FABP4 appeared to be associated with total mortality differentially in men and women (HR 1.17, 95% CI 1.08 to 1.26 for men; HR 1.02, 95% CI 0.96 to 1.07 for women, interaction p value <0.001). In conclusion, in a cohort of community-dwelling older subjects, elevated plasma concentrations of FFA, but not FABP4, were associated with cardiovascular and noncardiovascular mortality.
Collapse
Affiliation(s)
- Michael D Miedema
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Boston Veterans Affairs Healthcare System, Boston, Massachusetts; Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Minneapolis, Minnesota.
| | - Marlena Maziarz
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, Washington
| | - Mary L Biggs
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, Washington
| | - Susan J Zieman
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Jorge R Kizer
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York; Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Joachim H Ix
- Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, California; Divisions of Nephrology and Preventive Medicine, University of California-San Diego, San Diego, California
| | - Dariush Mozaffarian
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Russell P Tracy
- Department of Pathology, Colchester Research Facility, University of Vermont, Colchester, Virginia
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology, University of Washington, Seattle, Washington; Department of Health Services, University of Washington, Seattle, Washington; Group Health Research Institute, Group Health Cooperative, Seattle, Washington
| | - David S Siscovick
- Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology, University of Washington, Seattle, Washington
| | | | - Luc Djousse
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Boston Veterans Affairs Healthcare System, Boston, Massachusetts
| |
Collapse
|
35
|
Harjes U, Bridges E, McIntyre A, Fielding BA, Harris AL. Fatty acid-binding protein 4, a point of convergence for angiogenic and metabolic signaling pathways in endothelial cells. J Biol Chem 2014; 289:23168-23176. [PMID: 24939870 PMCID: PMC4132814 DOI: 10.1074/jbc.m114.576512] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fatty acid-binding protein 4 (FABP4) is an adipogenic protein and is implicated in atherosclerosis, insulin resistance, and cancer. In endothelial cells, FABP4 is induced by VEGFA, and inhibition of FABP4 blocks most of the VEGFA effects. We investigated the DLL4-NOTCH-dependent regulation of FABP4 in human umbilical vein endothelial cells by gene/protein expression and interaction analyses following inhibitor treatment and RNA interference. We found that FABP4 is directly induced by NOTCH. Stimulation of NOTCH signaling with human recombinant DLL4 led to FABP4 induction, independently of VEGFA. FABP4 induction by VEGFA was reduced by blockade of DLL4 binding to NOTCH or inhibition of NOTCH signal transduction. Chromatin immunoprecipitation of the NOTCH intracellular domain showed increased binding to two specific regions in the FABP4 promoter. The induction of FABP4 gene expression was dependent on the transcription factor FOXO1, which was essential for basal expression of FABP4, and FABP4 up-regulation following stimulation of the VEGFA and/or the NOTCH pathway. Thus, we show that the DLL4-NOTCH pathway mediates endothelial FABP4 expression. This indicates that induction of the angiogenesis-restricting DLL4-NOTCH can have pro-angiogenic effects via this pathway. It also provides a link between DLL4-NOTCH and FOXO1-mediated regulation of endothelial gene transcription, and it shows that DLL4-NOTCH is a nodal point in the integration of pro-angiogenic and metabolic signaling in endothelial cells. This may be crucial for angiogenesis in the tumor environment.
Collapse
Affiliation(s)
- Ulrike Harjes
- Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, OX3 9DS Oxford
| | - Esther Bridges
- Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, OX3 9DS Oxford
| | - Alan McIntyre
- Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, OX3 9DS Oxford
| | - Barbara A Fielding
- Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, and; Department of Nutritional Sciences, University of Surrey, GU2 7WG Surrey, United Kingdom
| | - Adrian L Harris
- Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, OX3 9DS Oxford,.
| |
Collapse
|
36
|
Saksi J, Ijäs P, Mäyränpää MI, Nuotio K, Isoviita PM, Tuimala J, Lehtonen-Smeds E, Kaste M, Jula A, Sinisalo J, Nieminen MS, Lokki ML, Perola M, Havulinna AS, Salomaa V, Kettunen J, Jauhiainen M, Kovanen PT, Lindsberg PJ. Low-expression variant of fatty acid-binding protein 4 favors reduced manifestations of atherosclerotic disease and increased plaque stability. ACTA ACUST UNITED AC 2014; 7:588-98. [PMID: 25122052 DOI: 10.1161/circgenetics.113.000499] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Fatty acid-binding protein 4 (FABP4 or aP2 in mice) has been identified as a key regulator of core aspects of cardiometabolic disorders, including lipotoxic endoplasmic reticulum stress in macrophages. A functional promoter polymorphism (rs77878271) of human FABP4 gene has been described resulting in reduced FABP4 transcription. METHODS AND RESULTS We investigated the effects of this low-expression variant of FABP4 on cardiovascular morbidity and carotid atherosclerosis on a population level (n=7491) and in patient cohorts representing endarterectomized patients with advanced carotid atherosclerosis (n=92) and myocardial infarction (n=3432). We found that the low-expression variant was associated with decreased total cholesterol levels (P=0.006) with the largest reduction in variant allele homozygotes. Obese variant allele carriers also showed reduced carotid intima-media thickness (P=0.010) and lower prevalence of carotid plaques (P=0.060). Consistently, the variant allele homozygotes showed 8-fold lower odds for myocardial infarction (P=0.019; odds ratio, 0.12; 95% confidence interval, 0.003-0.801). Within the carotid plaques, the variant allele was associated with a 3.8-fold reduction in FABP4 transcription (P=0.049) and 2.7-fold reduction in apoptosis (activated caspase 3; P=0.043). Furthermore, the variant allele was enriched to patients with asymptomatic carotid stenosis (P=0.038). High FABP4 expression in the carotid plaques was associated with lipid accumulation, intraplaque hemorrhages, plaque ulcerations, and phosphoactivated endoplasmic reticulum stress markers. CONCLUSIONS Our results reveal FABP4 rs77878271 as a novel variant affecting serum total cholesterol levels and cardiovascular risk. A therapeutic regimen reducing FABP4 expression within the atherosclerotic plaque may promote lesion stability through modulation of endoplasmic reticulum stress signaling, and attenuation of apoptosis, lipid burden, and inflammation.
Collapse
Affiliation(s)
- Jani Saksi
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.).
| | - Petra Ijäs
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Mikko I Mäyränpää
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Krista Nuotio
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Pia M Isoviita
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Jarno Tuimala
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Erno Lehtonen-Smeds
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Markku Kaste
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Antti Jula
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Juha Sinisalo
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Markku S Nieminen
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Marja-Liisa Lokki
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Markus Perola
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Aki S Havulinna
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Veikko Salomaa
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Johannes Kettunen
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Matti Jauhiainen
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Petri T Kovanen
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| | - Perttu J Lindsberg
- From the Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland (J.S., P.I., K.N., P.M.I., P.J.L.); HUSLAB, Division of Pathology (M.I.M.), Division of Cardiology, Department of Medicine (J.S., M.S.N.), Department of Neurology (P.I., K.N., M.K., P.J.L.), Helsinki University Central Hospital, Helsinki, Finland; Department of Pathology (M.I.M.), Transplantation Laboratory (M.-L.L.), Haartman Institute, Helsinki University, Helsinki, Finland; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland (M.P., J.K.); Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland (P.J.L.); Finnish Red Cross Blood Service, Helsinki, Finland (J.T.); Wihuri Research Institute, Helsinki, Finland (E.L.-S., P.T.K.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland (A.J.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (A.J., M.P., A.S.H., V.S., J.K., M.J.); and The Estonian Genome Center, University of Tartu, Tartu, Estonia (M.P.)
| |
Collapse
|
37
|
Okamoto R, Gery S, Gombart AF, Wang X, Castellani LW, Akagi T, Chen S, Arditi M, Ho Q, Lusis AJ, Li Q, Koeffler HP. Deficiency of CCAAT/enhancer binding protein-epsilon reduces atherosclerotic lesions in LDLR-/- mice. PLoS One 2014; 9:e85341. [PMID: 24489659 PMCID: PMC3904867 DOI: 10.1371/journal.pone.0085341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 12/04/2013] [Indexed: 01/23/2023] Open
Abstract
The CCAAT/enhancer binding proteins (C/EBPs) are transcription factors involved in hematopoietic cell development and induction of several inflammatory mediators. C/EBPε is expressed only in myeloid cells including monocytes/macrophages. Atherosclerosis is an inflammatory disorder of the vascular wall and circulating immune cells such as monocytes/macrophages. Mice deficient in the low density lipoprotein (LDL) receptor (Ldlr−/−) fed on a high cholesterol diet (HCD) show elevated blood cholesterol levels and are widely used as models to study human atherosclerosis. In this study, we generated Ldlr and Cebpe double-knockout (llee) mice and compared their atherogenic phenotypes to Ldlr single deficient (llEE) mice after HCD. Macrophages from llee mice have reduced lipid uptake by foam cells and impaired phagokinetic motility in vitro compared to macrophages from llEE mice. Also, compared to llEE mice, llee mice have alterations of lipid metabolism, and reduced atheroma and obesity, particularly the males. Peritoneal macrophages of llee male mice have reduced mRNA expression of FABP4, a fatty acid binding protein implicated in atherosclerosis. Overall, our study suggests that the myeloid specific factor C/EBPε is involved in systemic lipid metabolism and that silencing of C/EBPε could decrease the development of atherosclerosis.
Collapse
Affiliation(s)
- Ryoko Okamoto
- Division of Hematology and Oncology, Cedars-Sinai Medical Center, University of California Los Angeles (UCLA) School of Medicine, Los Angeles, California, United States of America
| | - Sigal Gery
- Division of Hematology and Oncology, Cedars-Sinai Medical Center, University of California Los Angeles (UCLA) School of Medicine, Los Angeles, California, United States of America
- * E-mail:
| | - Adrian F. Gombart
- Division of Hematology and Oncology, Cedars-Sinai Medical Center, University of California Los Angeles (UCLA) School of Medicine, Los Angeles, California, United States of America
- Department of Biochemisty and Biophysics, Linus Pauling Institute, Oregon State University, Corvallis, Oregon, United States of America
| | - Xuping Wang
- Department of Human Genetics, Department of Medicine, and Department of Microbiology, Molecular Genetics, and Immunology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, United States of America
| | - Lawrence W. Castellani
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, United States of America
| | - Tadayuki Akagi
- Division of Hematology and Oncology, Cedars-Sinai Medical Center, University of California Los Angeles (UCLA) School of Medicine, Los Angeles, California, United States of America
| | - Shuang Chen
- Division of Pediatric Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Moshe Arditi
- Division of Pediatric Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Quoc Ho
- Division of Hematology and Oncology, Cedars-Sinai Medical Center, University of California Los Angeles (UCLA) School of Medicine, Los Angeles, California, United States of America
| | - Aldons J. Lusis
- Department of Human Genetics, Department of Medicine, and Department of Microbiology, Molecular Genetics, and Immunology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California, United States of America
| | - Quanlin Li
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - H. Phillip Koeffler
- Division of Hematology and Oncology, Cedars-Sinai Medical Center, University of California Los Angeles (UCLA) School of Medicine, Los Angeles, California, United States of America
- Cancer Science Institute of Singapore and National Cancer Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
38
|
Parra S, Cabré A, Marimon F, Ferré R, Ribalta J, Gonzàlez M, Heras M, Castro A, Masana L. Circulating FABP4 is a marker of metabolic and cardiovascular risk in SLE patients. Lupus 2014; 23:245-54. [DOI: 10.1177/0961203313517405] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The aim of this study is to determine if circulating fatty acid-binding protein 4 (FABP4) plasma levels are a possible marker of metabolic risk in SLE patients. Circulating levels of adipose FABP4 are associated with adiposity, insulin resistance (IR), metabolic syndrome, diabetes and cardiovascular diseases. Patients affected by systemic lupus erythematosus (SLE) show an accelerated atherosclerosis that cannot be entirely explained by traditional cardiovascular risk factors. Sixty consecutive patients with SLE and 34 non-SLE age-matched controls were recruited for the study. Total plasma lipids and circulating FABP4 were determined. Subclinical atherosclerosis was evaluated by measuring carotid intimae-media thickness (c-IMT) by sonography, and the distribution of lipoprotein subclasses was analysed by nuclear magnetic resonance (NMR) spectroscopy. In the SLE group, FABP4 was associated with IR, atherogenic dyslipidaemia, as measured by NMR, and the presence of subclinical atherosclerosis. In multivariate analyses FABP4 was associated with increased c-IMT independent of the inflammatory state of the patient. In sum, circulating FABP4 is involved in the metabolic disturbances of SLE affecting lipid metabolism and IR, and it could be a biomarker of atherosclerosis in this population.
Collapse
Affiliation(s)
- S Parra
- Internal Medicine Department, “Sant Joan” University Hospital (Reus-Spain), Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
- Grupo de Enfermedades Autoinmunes Sistémicas (GEAS), Sociedad Española de Medicina Interna (SEMI), Spain
| | - A Cabré
- URLA, CIBERDEM, “Sant Joan” University Hospital (Reus-Spain), IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - F Marimon
- Internal Medicine Department, “Sant Joan” University Hospital (Reus-Spain), Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - R Ferré
- Internal Medicine Department, “Sant Joan” University Hospital (Reus-Spain), Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
- Unitat de Medicina Vascular i Metabolisme (UVASMET), Unitat de Recerca de Lipids i Arteriosclerosis (URLA), “Sant Joan” University Hospital (Reus-Spain), Internal Medicine, IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - J Ribalta
- URLA, CIBERDEM, “Sant Joan” University Hospital (Reus-Spain), IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - M Gonzàlez
- URLA, CIBERDEM, “Sant Joan” University Hospital (Reus-Spain), IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - M Heras
- URLA, CIBERDEM, “Sant Joan” University Hospital (Reus-Spain), IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - A Castro
- Internal Medicine Department, “Sant Joan” University Hospital (Reus-Spain), Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
- Grupo de Enfermedades Autoinmunes Sistémicas (GEAS), Sociedad Española de Medicina Interna (SEMI), Spain
| | - L Masana
- Internal Medicine Department, “Sant Joan” University Hospital (Reus-Spain), Institut Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
- Unitat de Medicina Vascular i Metabolisme (UVASMET), Unitat de Recerca de Lipids i Arteriosclerosis (URLA), “Sant Joan” University Hospital (Reus-Spain), Internal Medicine, IISPV, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
39
|
Valanti E, Tsompanidis A, Sanoudou D. Pharmacogenomics in the development and characterization of atheroprotective drugs. Methods Mol Biol 2014; 1175:259-300. [PMID: 25150873 DOI: 10.1007/978-1-4939-0956-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Atherosclerosis is the main cause of cardiovascular disease (CVD) and can lead to stroke, myocardial infarction, and death. The clinically available atheroprotective drugs aim mainly at reducing the levels of circulating low-density lipoprotein (LDL), increasing high-density lipoprotein (HDL), and attenuating inflammation. However, the cardiovascular risk remains high, along with morbidity, mortality, and incidence of adverse drug events. Pharmacogenomics is increasingly contributing towards the characterization of existing atheroprotective drugs, the evaluation of novel ones, and the identification of promising, unexplored therapeutic targets, at the global molecular pathway level. This chapter presents highlights of pharmacogenomics investigations and discoveries that have contributed towards the elucidation of pharmacological atheroprotection, while opening the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Efi Valanti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens, 115 27, Greece
| | | | | |
Collapse
|
40
|
Girona J, Rosales R, Plana N, Saavedra P, Masana L, Vallvé JC. FABP4 induces vascular smooth muscle cell proliferation and migration through a MAPK-dependent pathway. PLoS One 2013; 8:e81914. [PMID: 24312381 PMCID: PMC3843707 DOI: 10.1371/journal.pone.0081914] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 10/28/2013] [Indexed: 01/03/2023] Open
Abstract
Purpose The migration and proliferation of vascular smooth muscle cells play crucial roles in the development of atherosclerotic lesions. This study examined the effects of fatty acid binding protein 4 (FABP4), an adipokine that is associated with cardiovascular risk, endothelial dysfunction and proinflammatory effects, on the migration and proliferation of human coronary artery smooth muscle cells (HCASMCs). Methods and Results A DNA 5-bromo-2′-deoxy-uridine (BrdU) incorporation assay indicated that FABP4 significantly induced the dose-dependent proliferation of HCASMCs with a maximum stimulatory effect at 120 ng/ml (13% vs. unstimulated cells, p<0.05). An anti-FABP4 antibody (40 ng/ml) significantly inhibited the induced cell proliferation, demonstrating the specificity of the FABP4 proliferative effect. FABP4 significantly induced HCASMC migration in a dose-dependent manner with an initial effect at 60 ng/ml (12% vs. unstimulated cells, p<0.05). Time-course studies demonstrated that FABP4 significantly increased cell migration compared with unstimulated cells from 4 h (23%vs. 17%, p<0.05) to 12 h (74%vs. 59%, p<0.05). Pretreatment with LY-294002 (5 µM) and PD98059 (10 µM) blocked the FABP4-induced proliferation and migration of HCASMCs, suggesting the activation of a kinase pathway. On a molecular level, we observed an up-regulation of the MAPK pathway without activation of Akt. We found that FABP4 induced the active forms of the nuclear transcription factors c-jun and c-myc, which are regulated by MAPK cascades, and increased the expression of the downstream genes cyclin D1 and MMP2, CCL2, and fibulin 4 and 5, which are involved in cell cycle regulation and cell migration. Conclusions These findings indicate a direct effect of FABP4 on the migration and proliferation of HCASMCs, suggesting a role for this adipokine in vascular remodelling. Taken together, these results demonstrate that the FABP4-induced DNA synthesis and cell migration are mediated primarily through a MAPK-dependent pathway that activates the transcription factors c-jun and c-myc in HCASMCs.
Collapse
Affiliation(s)
- Josefa Girona
- Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Universitat Rovira i Virgili, IISPV, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Reus, Spain
| | - Roser Rosales
- Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Universitat Rovira i Virgili, IISPV, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Reus, Spain
| | - Núria Plana
- Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Universitat Rovira i Virgili, IISPV, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Reus, Spain
| | - Paula Saavedra
- Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Universitat Rovira i Virgili, IISPV, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Reus, Spain
| | - Lluís Masana
- Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Universitat Rovira i Virgili, IISPV, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Reus, Spain
| | - Joan-Carles Vallvé
- Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Universitat Rovira i Virgili, IISPV, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Reus, Spain
- * E-mail:
| |
Collapse
|
41
|
IL-1β, RAGE and FABP4: targeting the dynamic trio in metabolic inflammation and related pathologies. Future Med Chem 2013; 5:1089-108. [PMID: 23795967 DOI: 10.4155/fmc.13.90] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Within the past decade, inflammatory and lipid mediators, such as IL-1β, FABP4 and RAGE, have emerged as important contributors to metabolic dysfunction. As growing experimental and clinical evidence continues to tie obesity-induced chronic inflammation with dysregulated lipid, insulin signaling and related pathologies, IL-1β, FABP4 and RAGE each are being independently implicated as culprits in these events. There are also convincing data that molecular pathways driven by these molecules are interconnected in exacerbating metabolic consequences of obesity. This article highlights the roles of IL-1β, FABP4 and RAGE in normal physiology as well as focusing specifically on their contribution to inflammation, insulin resistance, atherosclerosis, Type 2 diabetes and cancer. Studies implicating the interconnection between these pathways, current and emerging therapeutics, and their use as potential biomarkers are also discussed. Evidence of impact of IL-1β, FABP4 and RAGE pathways on severity of metabolic dysfunction underlines the strong links between inflammatory events, lipid metabolism and insulin regulation, and offers new intriguing approaches for future therapies of obesity-driven pathologies.
Collapse
|
42
|
The level of fatty acid-binding protein 4, a novel adipokine, is increased in rheumatoid arthritis and correlates with serum cholesterol levels. Cytokine 2013; 64:441-7. [DOI: 10.1016/j.cyto.2013.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/26/2013] [Accepted: 05/01/2013] [Indexed: 01/01/2023]
|
43
|
Huang CL, Wu YW, Wu CC, Lin L, Wu YC, Hsu PY, Jong YS, Yang WS. Association between serum adipocyte fatty-acid binding protein concentrations, left ventricular function and myocardial perfusion abnormalities in patients with coronary artery disease. Cardiovasc Diabetol 2013; 12:105. [PMID: 23866022 PMCID: PMC3733628 DOI: 10.1186/1475-2840-12-105] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 07/15/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Adipokines, including adipocyte fatty acid-binding protein (A-FABP), have been demonstrated to be involved in the pathogenesis of atherosclerosis. In the present study, we investigated the association of circulating A-FABP level with severity of myocardial perfusion abnormalities analyzed by Tl-201 dipyridamole single-photon emission computed tomography. METHODS A total of 170 patients with coronary artery disease (CAD) from cardiovascular clinics were enrolled in the study. Serum A-FABP levels, echocardiography, and stress myocardial perfusion imaging results were analyzed. RESULTS Compared with the patients with mild CAD (summed stress score [SSS] ≤ 8), those with moderate to severe CAD (SSS > 8) had significantly higher A-FABP concentrations. However, the difference was attenuated in the subgroup of patients with heart failure. In the correlation analyses, A-FABP level was correlated with age, body mass index, waist circumference, levels of creatinine, fasting glucose, high-sensitivity C-reactive protein, N-terminal pro-brain natriuretic peptide, adiponectin, and several echocardiographic parameters, including left ventricular ejection fraction. Multivariate logistic regression analysis demonstrated that the A-FABP level was not only associated with higher SSS (odds ratio, 1.30; 95% confidence interval [CI], 1.01-1.69; P = 0.048), but also an independent risk factor for heart failure (odds ratio 2.71, 95% CI, 1.23-5.94; P = 0.013). CONCLUSIONS Serum A-FABP levels not only were associated with myocardial perfusion abnormalities and left ventricular function, but also predicted the presence of heart failure in our patients with CAD.
Collapse
Affiliation(s)
- Chi-Lun Huang
- Department of Internal Medicine, Taoyuan General Hospital, Taoyuan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Beniyama Y, Matsuno K, Miyachi H. Structure-guided design, synthesis and in vitro evaluation of a series of pyrazole-based fatty acid binding protein (FABP) 3 ligands. Bioorg Med Chem Lett 2013; 23:1662-6. [PMID: 23395658 DOI: 10.1016/j.bmcl.2013.01.054] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 01/11/2013] [Accepted: 01/16/2013] [Indexed: 12/01/2022]
Abstract
We designed a series of pyrazole-based carboxylic acids as candidate ligands of heart fatty acid binding protein (H-FABP, or FABP3), based on a comparison of the X-ray crystallographic structures of adipocyte fatty acid binding protein (FABP4)-selective inhibitor (BMS309403) complex and FABP3-elaidic acid complex. Some of the synthesized compounds exhibited dual FABP3/4 ligand activity, and some exhibited selectivity for FABP3.
Collapse
Affiliation(s)
- Yoko Beniyama
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | | | | |
Collapse
|
45
|
Kralisch S, Fasshauer M. Adipocyte fatty acid binding protein: a novel adipokine involved in the pathogenesis of metabolic and vascular disease? Diabetologia 2013; 56:10-21. [PMID: 23052058 DOI: 10.1007/s00125-012-2737-4] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 09/10/2012] [Indexed: 12/18/2022]
Abstract
Adipocyte fatty acid binding protein (AFABP, also known as aP2 and FABP4) has recently been introduced as a novel fat-derived circulating protein. AFABP serum concentrations are positively correlated with markers of the metabolic syndrome and vascular disease in various cross-sectional and interventional studies. Furthermore, a small set of prospective studies indicates that high AFABP serum levels at baseline predict the risk for metabolic and vascular morbidity and mortality. Studies in Afabp (also known as Fabp4) knockout mice and AFABP inhibitor-treated animals suggest that total AFABP promotes insulin resistance, hypertriacylglycerolaemia and atherosclerosis by ligand/ligand delivery, as well as ligand-independent mechanisms. In contrast, the pathophysiological significance of circulating AFABP and the mechanisms leading to its release remain to be established. The current review summarises recent findings on the regulation and potential role of AFABP in metabolic and vascular disease.
Collapse
Affiliation(s)
- S Kralisch
- Department of Endocrinology and Nephrology, University of Leipzig, Liebigstr 18, 04103 Leipzig, Germany
| | | |
Collapse
|
46
|
Lee K, Santibanez-Koref M, Polvikoski T, Birchall D, Mendelow AD, Keavney B. Increased expression of fatty acid binding protein 4 and leptin in resident macrophages characterises atherosclerotic plaque rupture. Atherosclerosis 2012; 226:74-81. [PMID: 23122912 PMCID: PMC3566542 DOI: 10.1016/j.atherosclerosis.2012.09.037] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 09/23/2012] [Accepted: 09/25/2012] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Resident macrophages play an important role in atheromatous plaque rupture. The macrophage gene expression signature associated with plaque rupture is incompletely defined due to the complex cellular heterogeneity in the plaque. We aimed to characterise differential gene expression in resident plaque macrophages from ruptured and stable human atheromatous lesions. METHODS AND RESULTS We performed genome-wide expression analyses of isolated macrophage-rich regions of stable and ruptured human atherosclerotic plaques. Plaques present in carotid endarterectomy specimens were designated as stable or ruptured using clinical, radiological and histopathological criteria. Macrophage-rich regions were excised from 5 ruptured and 6 stable plaques by laser micro-dissection. Transcriptional profiling was performed using Affymetrix microarrays. The profiles were characteristic of activated macrophages. At a false discovery rate of 10%, 914 genes were differentially expressed between stable and ruptured plaques. The findings were confirmed in fourteen further stable and ruptured samples for a subset of eleven genes with the highest expression differences (p < 0.05). Pathway analysis revealed that components of the PPAR/Adipocytokine signaling pathway were the most significantly upregulated in ruptured compared to stable plaques (p = 5.4 × 10(-7)). Two key components of the pathway, fatty-acid binding-protein 4 (FABP4) and leptin, showed nine-fold (p = 0.0086) and five-fold (p = 0.0012) greater expression respectively in macrophages from ruptured plaques. CONCLUSIONS We found differences in gene expression signatures between macrophages isolated from stable and ruptured human atheromatous plaques. Our findings indicate the involvement of FABP4 and leptin in the progression of atherosclerosis and plaque rupture, and suggest that down-regulation of PPAR/adipocytokine signaling within plaques may have therapeutic potential.
Collapse
Affiliation(s)
- K Lee
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, NE1 3BZ Newcastle-upon-Tyne, UK.
| | | | | | | | | | | |
Collapse
|